AU2013255612B2 - Pyrrolobenzodiazepines - Google Patents

Pyrrolobenzodiazepines Download PDF

Info

Publication number
AU2013255612B2
AU2013255612B2 AU2013255612A AU2013255612A AU2013255612B2 AU 2013255612 B2 AU2013255612 B2 AU 2013255612B2 AU 2013255612 A AU2013255612 A AU 2013255612A AU 2013255612 A AU2013255612 A AU 2013255612A AU 2013255612 B2 AU2013255612 B2 AU 2013255612B2
Authority
AU
Australia
Prior art keywords
compound according
nme
group
methyl
phenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2013255612A
Other versions
AU2013255612A1 (en
Inventor
Philip Wilson Howard
Khondaker Mirazur RAHMAN
Peter William Taylor
David Edwin Thurston
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune Ltd
Original Assignee
MedImmune Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by MedImmune Ltd filed Critical MedImmune Ltd
Publication of AU2013255612A1 publication Critical patent/AU2013255612A1/en
Assigned to UCL BUSINESS PLC, MEDIMMUNE LIMITED reassignment UCL BUSINESS PLC Request for Assignment Assignors: SPIROGEN SARL, UCL BUSINESS PLC
Assigned to MEDIMMUNE LIMITED reassignment MEDIMMUNE LIMITED Request for Assignment Assignors: MEDIMMUNE LIMITED, UCL BUSINESS PLC
Application granted granted Critical
Publication of AU2013255612B2 publication Critical patent/AU2013255612B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Oncology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

A compound of formula (I) or a salt or solvate thereof, wherein the dotted double bond indicates the presence of a single or double bond between C2 and C3; R

Description

PYRROLOBENZODIAZEPINES
The present invention relates to pyrrolobenzodiazepines (PBDs) and in particular to PBD monomers and methods of synthesising PBD monomers.
Background to the invention
Some pyrrolobenzodiazepines (PBDs) have the ability to recognise and bond to specific sequences of DNA; the preferred sequence is PuGPu. The first PBD antitumour antibiotic, anthramycin, was discovered in 1965 (Leimgruber, etal., J. Am. Chem. Soc., 87, 5793-5795 (1965); Leimgruber, etal., J. Am. Chem. Soc., 87, 5791-5793 (1965)). Since then, a number of naturally occurring PBDs have been reported, and over 10 synthetic routes have been developed to a variety of analogues (Thurston, et al., Chem. Rev. 1994, 433-465 (1994)). Family members include abbeymycin (Hochlowski, etal., J. Antibiotics, 40, 145-148 (1987)), chicamycin (Konishi, etal., J. Antibiotics, 37, 200-206 (1984)), DC-81 (Japanese Patent 58-180 487; Thurston, etal., Chem. Brit., 26, 767-772 (1990); Bose, etal., Tetrahedron, 48, 751-758 (1992)), mazethramycin (Kuminoto, etal., J. Antibiotics, 33, 665-667 (1980)), neothramycins A and B (Takeuchi, etal., J. Antibiotics, 29, 93-96 (1976)), porothramycin (Tsunakawa, etal., J. Antibiotics, 41, 1366-1373 (1988)), prothracarcin (Shimizu, etal, J. Antibiotics, 35, 972-978 (1982); Langley and Thurston, J. Org. Chem., 52, 91-97 (1987)), sibanomicin (DC-102)(Hara, etal., J. Antibiotics, 41, 702-704 (1988); Itoh, etal., J. Antibiotics, 41, 1281-1284 (1988)), sibiromycin (Leber, etal., J. Am. Chem. Soc., 110, 2992-2993 (1988)) and tomamycin (Arima, etal., J. Antibiotics, 25, 437-444 (1972)). PBDs are of the general structure:
They differ in the number, type and position of substituents, in both their aromatic A rings and pyrrolo C rings, and in the degree of saturation of the C ring. In the B-ring there is either an imine (N=C), a carbinolamine(NH-CH(OH)), or a carbinolamine methyl ether (NH-CH(OMe)) at the N10-C11 position which is the electrophilic centre responsible for alkylating DNA. All of the known natural products have an (^-configuration at the chiral C11a position which provides them with a right-handed twist when viewed from the C ring towards the A ring. This gives them the appropriate three-dimensional shape for isohelicity with the minor groove of B-form DNA, leading to a snug fit at the binding site (Kohn, In Antibiotics III. Springer-Verlag, New York, pp. 3-11 (1975); Hurley and Needham-VanDevanter, Acc. Chem. Res., 19, 230-237 (1986)). Their ability to form an adduct in the minor groove, enables them to interfere with DNA processing, hence their use as antitumour agents. The synthesis of the compounds has been reviewed in Thurston, D.E., etal., Chem. Rev., 1994, 94, 433-465 and Thurston, D.E., etal., Chem. Rev., 2011, 111, 2815-2864.
The large majority of antibiotics in current clinical use are derived from bactericidal or bacteriostatic molecules produced as secondary metabolites by microbes, predominantly those belonging to the moulds and actinobacteria. Screening has identified molecules such as the pyrrolobenzodiazepines (PBDs) tomaymycin, anthramycin and DC-81 that exerted potent antibacterial activity against human pathogens through a capacity to bind to DNA; some of these compounds have potential as cancer chemotherapeutics but a high degree of cytotoxicity has rendered them unattractive as antibacterial antibiotics in comparison to other classes of compound.
However, the evolution of multidrug-resistant pathogens capable of rapid and efficient horizontal transmission of genes encoding antibiotic resistance determinants has facilitated the erosion in a relatively short timeframe of much of the therapeutic value of frontline antibacterial chemotherapeutic agents. Increasing multidrug resistance in Gram-negative pathogens such as Pseudomonas aeruginosa and Acinetobacter baumannii has forced the reappraisal of colistin for systemic use; this polymyxin antibiotic, discovered more than fifty years ago and until recently considered too toxic for non-topical use, is now widely used systemically due to the limited therapeutic options available for these infections. A number of conjugates of PBD with pyrroles and imidazoles have been reported, such as:
where n = 1-3 (Damayanthi, Y., etal., Journal of Organic Chemistry, 64(1), 290-292 (1999));
where n=1-3 and
where n=1-2 (Kumar, R. and Lown, J.W. Oncology Research, 13(4), 221-233 (2003)); Kumar, R., et al., Heterocyclic Communications, 8(1), 19-26 (2002));
where n = 1-4, (Baraldi, P.G., etal., Journal of Medicinal Chemistry, 42(25), 5131-5141 (1999));
where n=3, (Wells, G., etal., Proc. Am. Assoc. Cane. Res., 2003, 44, 452).
In WO 2005/085177, some of the present inventors disclosed amino acids comprising a biaryl core that could have useful properties in DNA binding.
The inventors have now discovered that the PBD conjugates of the prior art may be modified in order to achieve improved properties, particularly antibacterial properties. In particular, the present invention relates to the incorporation of an amino acide residue containing a 5-mebered heterocyclic group in combination with an arylene based amino acid residue in a PBD conjugate results in highly effective compounds. A first aspect of the present invention provides a compound of formula I:
or a salt or solvate thereof, wherein: the dotted double bond indicates the presence of a single or double bond between C2 and C3; R2 is selected from -H, -OH, =0, =CH2, -CN, -R, OR, halo, dihalo, =CHR, =CHRR’, -0-S02-R, C02R and COR; R7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR’, nitro, Me3Sn and halo; where R and R’ are independently selected from optionally substituted C1.7 alkyl, C3.20 heterocyclyl and C5.2o aryl groups; R10 and R11 either together form a double bond, or are selected from H and QRq respectively, where Q is selected from 0, S and NH and RQ is H or C1.7 alkyl or H and SOxM, where x is 2 or 3, and M is a monovalent pharmaceutically acceptable cation; A is selected from A1, A2, A3, A4 or A5:
Where X1 and Y1 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and O, respectively; X2 and Y2 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and 0, respectively; Z1 is selected from 0 and S; Z2 is selected from CH and N; F is selected from a single bond and —(E-F1)m-; each E is independently selected from a single bond, and -C(=0)-NH-; each F1 is independently a C3.20 heteroarylene group; m is 1, 2 or 3; G is selected from hydrogen, Ci_4alkyl, -C(=0)-0-Ci.4alkyl, -(CH2)n-C3-2o heterocycloalkyl, and -0-(CH2) n-C3-2o heterocycloalkyl group, and each n is 0-4; provided that A2 is not A2’:
where X1 and Y1 of A2’ are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; and provided that A3 is not A3’:
where X2 and Y2 of A3’ are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; B in A2’ and A3’ is either a single bond or:
where X and Y of B1 are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; and R1 is Cm alkyl. A second aspect of the present invention provides a method of synthesis of a compound of formula I. A third aspect of the present invention provides a pharmaceutical composition comprising a compound of the first aspect of the invention and a pharmaceutically acceptable carrier or diluent. A fourth aspect of the present invention provides a compound of the first aspect of the invention for use in a method of therapy. A fifth aspect of the present invention provides the use of a compound of the first aspect of the invention in the manufacture of a medicament for the treatment of a bacterial infection. This aspect also provides a compound of the first aspect for use in a method of treatment of a bacterial infection. A sixth aspect of the present invention provides a method of treatment of a patient suffering from a bacterial infection, comprising administering to said patient a therapeutically acceptable amount of a compound of the first aspect or a composition of the third aspect.
In the fourth to sixth aspects of the invention, the compound of the invention may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. In the third aspect of the invention, the pharmaceutical composition may comprise one or more (e g. two , three or four) further active agents. A seventh aspect of the present invention provides a compound of formula I:
or a salt or solvate thereof, wherein: the dotted double bond indicates the presence of a single or double bond between C2 and C3; R2 is selected from -H; =CH2: =CH(Me); C5 20 aryl optionally substituted by 1 to 3 groups selected from methoxy, ethoxy, fluoro, chloro, cyano, 5 bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thienyl; C-|_5 saturated aliphatic alkyl; C3_6 saturated cycloalkyl;
, wherein each of R21, R22 and R23 are independently selected from H, C^3 saturated alkyl, C2_3 alkenyl, C2_3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R2 group is no more than 5;
, wherein one of R25a and R25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl and methoxy; pyridyl; and thiophenyl; and
where R24 is selected from: H; C13 saturated alkyl; C2_3 alkenyl; C2.3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl and methoxy; pyridyl; and thiophenyl; R7 is OR7A wherein R7Ais selected from Me, ChtePh and allyl; R10 and R11 either together form a double bond, or are selected from H and QR° respectively, where Q is selected from O, S and NH and RQ is H or C·,_7 alkyl or H and SOxM, where x is 2 or 3, and M is a monovalent pharmaceutically acceptable cation; A is selected from A1, A2, A3, A4 or A5:
where X1 and Y1 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and 0, respectively; X2 and Y2 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and 0, respectively; Z1 is selected from O and S; Z2 is selected from CH and N; F is selected from a single bond and -(E-F1)m-; each E is independently selected from a single bond, and -C(=0)-NFI-; each F1 is independently a C3.2o heteroarylene group; m is 1,2 or 3; G is selected from hydrogen, C14alkyl, -C(=0)-0-C1_4alkyl, -(CFl2)n-C3-2o heterocycloalkyl, and -0-(CH2)n_C3.2o heterocycloalkyl group; each n is 0-4; provided that A2 is not A2’:
where X1 and Y1 of A2’ are selected from: CFI and NMe; COFI and NMe; CH and S; N and NMe; N and S, respectively; and provided that A3 is not A3’:
where X2 and Y2 of A3’ are selected from: CFI and NMe; COFI and NMe; CFI and S; N and NMe; N and S, respectively; B is either a single bond or:
where X and Y of B1 are selected from: CFI and NMe; COFt and NMe; CFI and S; N and NMe; N and S, respectively; and R1 is C-|_4 alkyl.
Definitions
Substituents
The phrase “optionally substituted” as used herein, pertains to a parent group which may be unsubstituted or which may be substituted.
Unless otherwise specified, the term “substituted” as used herein, pertains to a parent group which bears one or more substitutents. The term “substituent” is used herein in the conventional sense and refers to a chemical moiety which is covalently attached to, or if appropriate, fused to, a parent group. A wide variety of substituents are well known, and methods for their formation and introduction into a variety of parent groups are also well known.
Examples of substituents are described in more detail below.
Ci_7 alkyl: The term “C-i_7 alkyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to 7 carbon atoms, which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated). Thus, the term “alkyl” includes the sub-classes alkenyl, alkynyl, cycloalkyl, etc., discussed below.
Examples of saturated alkyl groups include, but are not limited to, methyl (Ci), ethyl (C2)> propyl (C3), butyl (C4), pentyl (C5), hexyl (C6) and heptyl (C7).
Examples of saturated linear alkyl groups include, but are not limited to, methyl (C1), ethyl (C2), n-propyl (C3), n-butyl (C4), n-pentyl (amyl) (C5), n-hexyl (C6) and n-heptyl (C7)·
Examples of saturated branched alkyl groups include iso-propyl (C3), iso-butyl (C4), sec-butyl (C4), tert-butyl (C4), iso-pentyl (C5), and neo-pentyl (C5). C2.7 Alkenyl: The term “C2-7 alkenyl” as used herein, pertains to an alkyl group having one or more carbon-carbon double bonds.
Examples of unsaturated alkenyl groups include, but are not limited to, ethenyl (vinyl, -CH=CH2), 1-propenyl (-CH=CH-CH3), 2-propenyl (allyl, -CH-CH=CH2), isopropenyl (1-methylvinyl, -C(CH3)=CH2), butenyl (C4), pentenyl (C5), and hexenyl (C6). C2.7 alkynyl: The term “C2-7 alkynyl” as used herein, pertains to an alkyl group having one or more carbon-carbon triple bonds.
Examples of unsaturated alkynyl groups include, but are not limited to, ethynyl (ethinyl, -C^CH) and 2-propynyl (propargyl, -CH2-CeCH). C3.7 cycloalkyl: The term “C3.7 cycloalkyl” as used herein, pertains to an alkyl group which is also a cyclyl group; that is, a monovalent moiety obtained by removing a hydrogen atom from an alicyclic ring atom of a cyclic hydrocarbon (carbocyclic) compound, which moiety has from 3 to 7 carbon atoms, including from 3 to 7 ring atoms.
Examples of cycloalkyl groups include, but are not limited to, those derived from: saturated monocyclic hydrocarbon compounds: cyclopropane (C3), cyclobutane (C4), cyclopentane (C5), cyclohexane (C6), cycloheptane (C7), methylcyclopropane (C4), dimethylcyclopropane (C5), methylcyclobutane (C5), dimethylcyclobutane (C6), methylcyclopentane (C6), dimethylcyclopentane (C7) and methylcyclohexane (C7); unsaturated monocyclic hydrocarbon compounds: cyclopropene (C3), cyclobutene (C4), cyclopentene (C5), cyclohexene (C6), methylcyclopropene (C4), dimethylcyclopropene (C5), methylcyclobutene (C5), dimethylcyclobutene (C6), methylcyclopentene (C6), dimethylcyclopentene (C7) and methylcyclohexene (C7); and saturated polycyclic hydrocarbon compounds: norcarane (C7), norpinane (C7), norbornane (C7). C3.2o heterocyclyl: The term “C3.2o heterocyclyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atoms from a ring atom of a heterocyclic compound, which moiety has from 3 to 20 ring atoms, of which from 1 to 10 are ring heteroatoms. Preferably, each ring has from 3 to 7 ring atoms, of which from 1 to 4 are ring heteroatoms.
In this context, the prefixes (e.g. C3-20, C3.7, C5-6, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term “Cs-eheterocyclyl”, as used herein, pertains to a heterocyclyl group having 5 or 6 ring atoms.
Examples of monocyclic heterocyclyl groups include, but are not limited to, those derived from:
Nv aziridine (C3), azetidine (C4), pyrrolidine (tetrahydropyrrole) (C5), pyrroline (e.g., 3-pyrroline, 2,5-dihydropyrrole) (C5), 2H-pyrrole or 3H-pyrrole (isopyrrole, isoazole) (C5), piperidine (C6), dihydropyridine (C6), tetrahydropyridine (C6), azepine (C7); 0{. oxirane (C3), oxetane (C4), oxolane (tetrahydrofuran) (C5), oxole (dihydrofuran) (C5), oxane (tetrahydropyran) (C6), dihydropyran (C6), pyran (C6), oxepin (C7);
Si: thiirane (C3), thietane (C4), thiolane (tetrahydrothiophene) (C5), thiane (tetrahydrothiopyran) (C6), thiepane (C7); 02: dioxolane (C5), dioxane (C6), and dioxepane (C7); 03: trioxane (C6); N2: imidazolidine (C5), pyrazolidine (diazolidine) (C5), imidazoline (C5), pyrazoline (dihydropyrazole) (C5), piperazine (C6); N1O1: tetrahydrooxazole (C5), dihydrooxazole (C5), tetrahydroisoxazole (C5), dihydroisoxazole (C5), morpholine (C6), tetrahydrooxazine (C6), dihydrooxazine (C6), oxazine (Ce); N1S1: thiazoline (C5), thiazolidine (C5), thiomorpholine (C6); N2Oi: oxadiazine (C6); O1S1: oxathiole (C5) and oxathiane (thioxane) (C6); and, N1O1S1: oxathiazine (C6).
Examples of substituted monocyclic heterocyclyl groups include those derived from saccharides, in cyclic form, for example, furanoses (C5), such as arabinofuranose, lyxofuranose, ribofuranose, and xylofuranse, and pyranoses (C6), such as allopyranose, altropyranose, glucopyranose, mannopyranose, gulopyranose, idopyranose, galactopyranose, and talopyranose.
Cs-2o aryl: The term “C5.2o aryl”, as used herein, pertains to a monovalent moiety obtained by removing from an aromatic ring atom of an aromatic compound, which moiety has from 3 to 20 ring atoms. Preferably, each ring has from 5 to 7 ring atoms.
In this context, the prefixes (e.g. C3-20, C5.7, C5-e, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term “C5.6 aryl” as used herein, pertains to an aryl group having 5 or 6 ring atoms.
The ring atoms may be all carbon atoms, as in “carboaryl groups”.
Examples of carboaryl groups include, but are not limited to, those derived from benzene (i.e. phenyl) (C6), naphthalene (C10), azulene (C10), anthracene (C14), phenanthrene (C14), naphthacene (Ci8), and pyrene (C16).
Examples of aryl groups which comprise fused rings, at least one of which is an aromatic ring, include, but are not limited to, groups derived from indane (e.g. 2,3-dihydro-1H-indene) (C9), indene (C9), isoindene (C9), tetraline (1,2,3,4-tetrahydronaphthalene (C10), acenaphthene (C12), fluorene (C13), phenalene (C13), acephenanthrene (C15), and aceanthrene (Ci6).
Alternatively, the ring atoms may include one or more heteroatoms, as in “heteroaryl groups”. Examples of monocyclic heteroaryl groups include, but are not limited to, those derived from:
Nh: pyrrole (azole) (C5), pyridine (azine) (C6); O1: furan (oxole) (C5);
Si: thiophene (thiole) (C5);
NiCh: oxazole (C5), isoxazole (C5), isoxazine (C6); N2Oi: oxadiazole (furazan) (C5); 1^3(^: oxatriazole (C5);
NiSii thiazole (C5), isothiazole (C5); N2: imidazole (1,3-diazole) (C5), pyrazole (1,2-diazole) (C5), pyridazine (1,2-diazine) (C6), pyrimidine (1,3-diazine) (C6) (e.g., cytosine, thymine, uracil), pyrazine (1,4-diazine) (C6); N3: triazole (C5), triazine (C6); and, N4: tetrazole (C5).
Examples of heteroaryl which comprise fused rings, include, but are not limited to: C9(with 2 fused rings) derived from benzofuran (Oi), isobenzofuran (Oi), indole (N^, isoindole (Ni), indolizine (N^, indoline (Ni), isoindoline (Ni), purine (N4) (e.g., adenine, guanine), benzimidazole (N2), indazole (N2), benzoxazole (N1O1), benzisoxazole (N1Oi), benzodioxole (02), benzofurazan (N2Oi), benzotriazole (N3), benzothiofuran (Si), benzothiazole (N^i), benzothiadiazole (N2S);
Cio(with 2 fused rings) derived from chromene (Oi), isochromene (Oi), chroman (0^, isochroman (Oi), benzodioxan (02), quinoline (N^, isoquinoline (N^, quinolizine (Ni), benzoxazine (N10i), benzodiazine (N2), pyridopyridine (N2), quinoxaline (N2), quinazoline (N2), cinnoline (N2), phthalazine (N2), naphthyridine (N2), pteridine (N4);
Cn (with 2 fused rings) derived from benzodiazepine (N2); C13(with 3 fused rings) derived from carbazole (NO, dibenzofuran (OO, dibenzothiophene (SO, carboline (N2), perimidine (N2), pyridoindole (N2); and, C14 (with 3 fused rings) derived from acridine (NO, xanthene (OO, thioxanthene (SO, oxanthrene (02), phenoxathiin (OiSO, phenazine (N2), phenoxazine (Ν·)Ο0, phenothiazine (NiSO, thianthrene (S2), phenanthridine (NO, phenanthroline (N2), phenazine (N2).
The terms “arylene”, “carboarylene”, “heteroarylene” as used herein, pertain to a divalent moiety obtained by removing a hydrogen atom from each of two ring atoms of a aromatic compound as described above with reference to aryl, carboaryl and heteroaryl.
The above groups, whether alone or part of another substituent, may themselves optionally be substituted with one or more groups selected from themselves and the additional substituents listed below.
Halo: -F, -Cl, -Br, and -I.
Hydroxy: -OH.
Ether: -OR, wherein R is an ether substituent, for example, a C^7 alkyl group (also referred to as a Ci_7 alkoxy group, discussed below), a C3.20 heterocyclyl group (also referred to as a C3.20 heterocyclyloxy group), or a C5.20 aryl group (also referred to as a C5.2oaryloxy group), preferably a C^alkyl group.
Alkoxy: -OR, wherein R is an alkyl group, for example, a C1.7 alkyl group. Examples of C1.7 alkoxy groups include, but are not limited to, -OMe (methoxy), -OEt (ethoxy), -O(nPr) (n-propoxy), -O(iPr) (isopropoxy), -O(nBu) (n-butoxy), -O(sBu) (sec-butoxy), -O(iBu) (isobutoxy), and -O(tBu) (tert-butoxy).
Acetal: -CH(OR1)(OR2), wherein R1 and R2 are independently acetal substituents, for example, a Ci.7alkyl group, a C3.2oheterocyclyl group, ora C5.2oaryl group, preferably a C1.7 alkyl group, or, in the case of a “cyclic” acetal group, R1 and R2, taken together with the two oxygen atoms to which they are attached, and the carbon atoms to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Examples of acetal groups include, but are not limited to, -CH(OMe)2, -CH(OEt)2, and -CH(OMe)(OEt).
Hemiacetal: -CH(OH)(OR1), wherein R1 is a hemiacetal substituent, for example, a 0V1 alkyl group, a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably a C1.7 alkyl group. Examples of hemiacetal groups include, but are not limited to, -CH(OH)(OMe) and -CH(OH)(OEt).
Ketal: -CR(OR1)(OR2), where R1 and R2 are as defined for acetals, and R is a ketal substituent other than hydrogen, for example, a Ci.7 alkyl group, a C3.20 heterocyclyl group, or a C5-2oaryl group, preferably a Ci.7 alkyl group. Examples ketal groups include, but are not limited to, -C(Me)(OMe)2, -C(Me)(OEt)2, -C(Me)(OMe)(OEt), -C(Et)(OMe)2, -C(Et)(OEt)2, and -C(Et)(OMe)(OEt).
Hemiketal: -CR(OH)(OR1), where R1 is as defined for hemiacetals, and R is a hemiketal substituent other than hydrogen, for example, a Ci_7 alkyl group, a C3.2o heterocyclyl group, or a C^aryl group, preferably a Ci_7 alkyl group. Examples of hemiacetal groups include, but are not limited to, -C(Me)(OH)(OMe), -C(Et)(OH)(OMe), -C(Me)(OH)(OEt), and -C(Et)(OH)(OEt).
Oxo (keto, -one): =0.
Thione (thioketone): =S.
Imino (imine): =NR, wherein R is an imino substituent, for example, hydrogen, alkyl group, a C3.20 heterocyclyl group, or a C5.2o aryl group, preferably hydrogen or a Ci.7 alkyl group. Examples of ester groups include, but are not limited to, =NH, =NMe, =NEt, and =NPh.
Formyl (carbaldehyde, carboxaldehyde): -C(=0)H.
Acyl (keto): -C(=0)R, wherein R is an acyl substituent, for example, a C1.7 alkyl group (also referred to as C1.7 alkylacyl or C1.7 alkanoyl), a C3.2o heterocyclyl group (also referred to as C3-20 heterocyclylacyl), or a C5.20 aryl group (also referred to as C5.20 arylacyl), preferably a C1.7 alkyl group. Examples of acyl groups include, but are not limited to, -C(=0)CH3 (acetyl), -C(=0)CH2CH3 (propionyl), -C(=0)C(CH3)3 (t-butyryl), and -C(=0)Ph (benzoyl, phenone).
Carboxy (carboxylic acid): -C(=0)0H.
Thiocarboxy (thiocarboxylic acid): -C(=S)SH.
Thiolocarboxy (thiolocarboxylic acid): -C(=0)SH.
Thionocarboxy (thionocarboxylic acid): -C(=S)OH.
Imidic acid: -C(=NH)OH.
Hydroxamic acid: -C(=NOH)OH.
Ester (carboxylate, carboxylic acid ester, oxycarbonyl): -C(=0)0R, wherein R is an ester substituent, for example, a C1.7 alkyl group, a C3.20 heterocyclyl group, or a C5.2o aryl group, preferably a C1.7 alkyl group. Examples of ester groups include, but are not limited to, -C(=0)0CH3, -C(=0)0CH2CH3, -C(=0)0C(CH3)3, and -C(=0)0Ph.
Acyloxy (reverse ester): -0C(=0)R, wherein R is an acyloxy substituent, for example, a C1.7 alkyl group, a C3.20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci_7 alkyl group. Examples of acyloxy groups include, but are not limited to, -0C(=0)CH3 (acetoxy), -0C(=0)CH2CH3, -0C(=0)C(CH3)3, -0C(=0)Ph, and -0C(=0)CH2Ph.
Oxycarboyloxy: -0C(=0)0R, wherein R is an ester substituent, for example, a alkyl group, a C3.20 heterocyclyl group, or a C5.2o aryl group, preferably a C1.7 alkyl group. Examples of ester groups include, but are not limited to, -0C(=0)0CH3, -0C(=0)0CH2CH3, -0C(=0)0C(CH3)3, and -0C(=0)0Ph.
Amino: -NR1R2, wherein R1 and R2 are independently amino substituents, for example, hydrogen, a Ci.7 alkyl group (also referred to as Ci.7alkylamino or di-Ci.7alkylamino), a C3.20 heterocyclyl group, or a C5-20 aryl group, preferably H or a Ci.7 alkyl group, or, in the case of a “cyclic” amino group, R1 and R2, taken together with the nitrogen atom to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Amino groups may be primary (-NH2), secondary (-NHR1), or tertiary (-NHR1R2), and in cationic form, may be quaternary (-+NR1R2R3). Examples of amino groups include, but are not limited to, -NH2, -NHCH3, -NHC(CH3)2, -N(CH3)2, -N(CH2CH3)2, and -NHPh. Examples of cyclic amino groups include, but are not limited to, aziridino, azetidino, pyrrolidino, piperidino, piperazino, morpholino, and thiomorpholino.
Amido (carbamoyl, carbamyl, aminocarbonyl, carboxamide): -C(=0)NR1R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=0)NH2, -C(=0)NHCH3, -C(=0)N(CH3)2, -C(=0)NHCH2CH3, and -C(=0)N(CH2CH3)2, as well as amido groups in which R1 and R2, together with the nitrogen atom to which they are attached, form a heterocyclic structure as in, for example, piperidinocarbonyl, morpholinocarbonyl, thiomorpholinocarbonyl, and piperazinocarbonyl.
Thioamido (thiocarbamyl): -C(=S)NR1R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=S)NH2, -C(=S)NHCH3, -C(=S)N(CH3)2, and -C(=S)NHCH2CH3.
Acylamido (acylamino): -NR1C(=0)R2, wherein R1 is an amide substituent, for example, hydrogen, a Ci.7alkyl group, a C3.20heterocyclyl group, ora C5.20aryl group, preferably hydrogen or a Ci.7 alkyl group, and R2 is an acyl substituent, for example, a Ci_7 alkyl group, a C3.2o heterocyclyl group, or a C5.2oaryl group, preferably hydrogen or a Ci_7 alkyl group. Examples of acylamide groups include, but are not limited to, -NHC(=0)CH3, -NHC(=0)CH2CH3, and -NHC(=0)Ph. R1 and R2 may together form a cyclic structure, as in, for example, succinimidyl, maleimidyl, and phthalimidyl:
Aminocarbonyloxy: -0C(=0)NR1R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of aminocarbonyloxy groups include, but are not limited to, -0C(=0)NH2, -0C(=0)NHMe, -0C(=0)NMe2, and -0C(=0)NEt2.
Ureido: -N(R1)CONR2R3 wherein R2 and R3 are independently amino substituents, as defined for amino groups, and R1 is a ureido substituent, for example, hydrogen, a C1.7 alkyl group, a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably hydrogen or a C1.7 alkyl group. Examples of ureido groups include, but are not limited to, -NHCONH2, -NHCONHMe, -NHCONHEt, -NHCONMe2, -NHCONEt2, -NMeCONH2, -NMeCONHMe, -NMeCONHEt, -NMeCONMe2, and -NMeCONEt2.
Guanidino: -NH-C(=NH)NH2.
Tetrazolyl: a five membered aromatic ring having four nitrogen atoms and one carbon atom,
Imino: =NR, wherein R is an imino substituent, for example, for example, hydrogen, a C1-7 alkyl group, a C3.20 heterocyclyl group, or a C5.20aryl group, preferably H or a Ci.7alkyl group. Examples of imino groups include, but are not limited to, =NH, =NMe, and =NEt.
Amidine (amidino): -C(=NR)NR2, wherein each R is an amidine substituent, for example, hydrogen, a C1.7 alkyl group, a C3.20 heterocyclyl group, or a C5-2oaryl group, preferably H or a C1.7 alkyl group. Examples of amidine groups include, but are not limited to, -C(=NH)NH2, -C(=NH)NMe2, and -C(=NMe)NMe2.
Nitro: -N02.
Nitroso: -NO.
Azido: -N3.
Cyano (nitrile, carbonitrile): -CN.
Isocyano: -NC.
Cyanato: -OCN.
Isocyanato: -NCO.
Thiocyano (thiocyanato): -SCN.
Isothiocyano (isothiocyanato): -NCS.
Sulfhydryl (thiol, mercapto): -SH.
Thioether (sulfide): -SR, wherein R is a thioether substituent, for example, a C1-7 alkyl group (also referred to as a Ci.7alkylthio group), a C3-20 heterocyclyl group, or a C5.2o aryl group, preferably a Ci_7 alkyl group. Examples of Ci_7 alkylthio groups include, but are not limited to, -SCH3 and -SCH2CH3.
Disulfide: -SS-R, wherein R is a disulfide substituent, for example, a Ci_7 alkyl group, a C3.20 heterocyclyl group, or a C5.2oaryl group, preferably a Ci.7 alkyl group (also referred to herein as Ci.7 alkyl disulfide). Examples of Ci_7 alkyl disulfide groups include, but are not limited to, -SSCH3 and -SSCH2CH3.
Sulfine (sulfinyl, sulfoxide): -S(=0)R, wherein R is a sulfine substituent, for example, a Ci.7alkyl group, a C3.20heterocyclyl group, ora C5-2oaryl group, preferably a Ci.7alkyl group. Examples of sulfine groups include, but are not limited to, -S(=0)CH3 and -S(=0)CH2CH3.
Sulfone (sulfonyl): -S(=0)2R, wherein R is a sulfone substituent, for example, a C^7 alkyl group, a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably a Ci.7 alkyl group, including, for example, a fluorinated or perfluorinated alkyl group. Examples of sulfone groups include, but are not limited to, -S(=0)2CH3 (methanesulfonyl, mesyl), -S(=0)2CF3 (triflyl), -S(=0)2CH2CH3 (esyl), -S(=0)2C4F9 (nonaflyl), -S(=0)2CH2CF3 (tresyl), -S(=0)2CH2CH2NH2 (tauryl), -S(=0)2Ph (phenylsulfonyl, besyl), 4-methylphenylsulfonyl (tosyl), 4-chlorophenylsulfonyl (closyl), 4-bromophenylsulfonyl (brosyl), 4-nitrophenyl (nosyl), 2-naphthalenesulfonate (napsyl), and 5-dimethylamino-naphthalen-1-ylsulfonate (dansyl).
Sulfinic acid (sulfino): -S(=0)0H, -S02H.
Sulfonic acid (sulfo): -S(=0)20H, -S03H.
Sulfinate (sulfinic acid ester): -S(=0)0R; wherein R is a sulfinate substituent, for example, a C1.7alkyl group, a C3.20heterocyclyl group, ora C5-20aryl group, preferably a Ci_7 alkyl group. Examples of sulfinate groups include, but are not limited to, -S(=0)0CH3 (methoxysulfinyl; methyl sulfinate) and -S(=0)0CH2CH3 (ethoxysulfinyl; ethyl sulfinate).
Sulfonate (sulfonic acid ester): -S(=0)20R, wherein R is a sulfonate substituent, for example, a C1.7 alkyl group, a C3.20 heterocyclyl group, or a C5-2oaryl group, preferably a C1.7 alkyl group. Examples of sulfonate groups include, but are not limited to, -S(=0)20CH3 (methoxysulfonyl; methyl sulfonate) and -S(=0)20CH2CH3 (ethoxysulfonyl; ethyl sulfonate).
Sulfinyloxy: -0S(=0)R, wherein R is a sulfinyloxy substituent, for example, a C1.7 alkyl group, a C3.20heterocyclyl group, ora C5.20aryl group, preferably a C1.7alkyl group. Examples of sulfinyloxy groups include, but are not limited to, -0S(=0)CH3 and -0S(=0)CH2CH3.
Sulfonyloxy: -0S(=0)2R, wherein R is a sulfonyloxy substituent, for example, a C1.7 alkyl group, a C3.20heterocyclyl group, ora C5.2oaryl group, preferably a C1.7alkyl group. Examples of sulfonyloxy groups include, but are not limited to, -0S(=0)2CH3 (mesylate) and -0S(=0)2CH2CH3 (esylate).
Sulfate: -0S(=0)20R; wherein R is a sulfate substituent, for example, a 0λ.7 alkyl group, a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably a C1.7 alkyl group. Examples of sulfate groups include, but are not limited to, -0S(=0)20CH3 and -S0(=0)20CH2CH3.
Sulfamyl (sulfamoyl; sulfinic acid amide; sulfinamide): -S(=0)NR1R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of sulfamyl groups include, but are not limited to, -S(=0)NH2, -S(=0)NH(CH3), -S(=0)N(CH3)2, -S(=0)NH(CH2CH3), -S(=0)N(CH2CH3)2, and -S(=0)NHPh.
Sulfonamido (sulfinamoyl; sulfonic acid amide; sulfonamide): -S(=0)2NR1R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of sulfonamido groups include, but are not limited to, -S(=0)2NH2, -S(=0)2NH(CH3), -S(=0)2N(CH3)2, -S(=0)2NH(CH2CH3), -S(=0)2N(CH2CH3)2, and -S(=0)2NHPh.
Sulfamino: -NR1S(=0)20H, wherein R1 is an amino substituent, as defined for amino groups. Examples of sulfamino groups include, but are not limited to, -NHS(=0)20H and -N(CH3)S(=0)20H.
Sulfonamino: -NR1S(=0)2R, wherein R1 is an amino substituent, as defined for amino groups, and R is a sulfonamino substituent, for example, a alkyl group, a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably a C1.7 alkyl group. Examples of sulfonamino groups include, but are not limited to, -NHS(=0)2CH3 and -N(CH3)S(=0)2C6H5.
Sulfinamino: -NR1S(=0)R, wherein R1 is an amino substituent, as defined for amino groups, and R is a sulfinamino substituent, for example, a C1.7 alkyl group, a C3.2o heterocyclyl group, or a C5.20 aryl group, preferably a C1.7 alkyl group. Examples of sulfinamino groups include, but are not limited to, -NHS(=0)CH3 and -N(CH3)S(=0)C6H5.
Phosphino (phosphine): -PR2, wherein R is a phosphino substituent, for example, -H, a C1-7 alkyl group, a C3.20 heterocyclyl group, or a C5-20 aryl group, preferably -H, a C1.7 alkyl group, or a C^aryl group. Examples of phosphino groups include, but are not limited to, -PH2, -P(CH3)2, -P(CH2CH3)2, -P(t-Bu)2, and -P(Ph)2.
Phospho: -P(=0)2.
Phosphinyl (phosphine oxide): -P(=0)R2, wherein R is a phosphinyl substituent, for example, a C1.7alkyl group, a C3.20heterocyclyl group, ora C5.20aryl group, preferably a C1.7 alkyl group or a C5.20aryl group. Examples of phosphinyl groups include, but are not limited to, -P(=0)(CH3)2, -P(=0)(CH2CH3)2, -P(=0)(t-Bu)2, and -P(=0)(Ph)2.
Phosphonic acid (phosphono): -P(=0)(0H)2.
Phosphonate (phosphono ester): -P(=0)(0R)2, where R is a phosphonate substituent, for example, -H, a Ci_7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably -H, a Ci_7 alkyl group, or a C5-20 aryl group. Examples of phosphonate groups include, but are not limited to, -P(=0)(0CH3)2, -P(=0)(0CH2CH3)2, -P(=0)(0-t-Bu)2, and -P(=0)(0Ph)2.
Phosphoric acid (phosphonooxy): -0P(=0)(0H)2.
Phosphate (phosphonooxy ester): -0P(=0)(0R)2, where R is a phosphate substituent, for example, -H, a Ci.7 alkyl group, a C3.20 heterocyclyl group, or a C5.20aryl group, preferably -H, a Ct.7 alkyl group, or a C5.20 aryl group. Examples of phosphate groups include, but are not limited to, -0P(=0)(0CH3)2, -0P(=0)(0CH2CH3)2, -0P(=0)(0-t-Bu)2, and -0P(=0)(0Ph)2.
Phosphorous acid: -OP(OH)2.
Phosphite: -OP(OR)2, where R is a phosphite substituent, for example, -H, a Ci.7 alkyl group, a C3.20 heterocyclyl group, or a C5-2oaryl group, preferably -H, a Ci_7 alkyl group, or a C5.20 aryl group. Examples of phosphite groups include, but are not limited to, -OP(OCH3)2, -OP(OCH2CH3)2, -0P(0-t-Bu)2, and -OP(OPh)2.
Phosphoramidite: -OP(OR1)-NR22, where R1 and R2 are phosphoramidite substituents, for example, -H, a (optionally substituted) Ci_7 alkyl group, a C3.20 heterocyclyl group, or a C5.2o aryl group, preferably -H, a Ci.7 alkyl group, or a C5.2o aryl group. Examples of phosphoramidite groups include, but are not limited to, -OP(OCH2CH3)-N(CH3)2, -OP(OCH2CH3)-N(i-Pr)2, and -OP(OCH2CH2CN)-N(i-Pr)2.
Phosphoramidate: -0P(=0)(0R1)-NR22, where R1 and R2 are phosphoramidate substituents, for example, -H, a (optionally substituted) alkyl group, a C3.20 heterocyclyl group, or a C5.20aryl group, preferably -H, a Ci.7 alkyl group, or a C5.20aryl group. Examples of phosphoramidate groups include, but are not limited to, -0P(=0)(0CH2CH3)-N(CH3)2, -0P(=0)(0CH2CH3)-N(i-Pr)2, and -0P(=0)(0CH2CH2CN)-N(i-Pr)2.
Nitrogen protecting groups
Nitrogen protecting groups are well known in the art. Preferred nitrogen protecting groups are carbamate protecting groups that have the general formula:
A large number of possible carbamate nitrogen protecting groups are listed on pages 706 to 771 of Wuts, P.G.M. and Greene, T.W., Protective Groups in Organic Synthesis, 4th Edition, Wiley-lnterscience, 2007, which is incorporated herein by reference.
Particularly preferred protecting groups include Alloc, Troc, Teoc, BOC, Doc, Hoc, TcBOC, Fmoc, 1-Adocand 2-Adoc.
Hydroxyl protecting groups
Hydroxyl protecting groups are well known in the art. A large number of suitable groups are described on pages 16 to 366 of Wuts, P.G.M. and Greene, T.W., Protective Groups in Organic Synthesis, 4th Edition, Wiley-lnterscience, 2007, which is incorporated herein by reference.
Classes of particular interest include silyl ethers, methyl ethers, alkyl ethers, benzyl ethers, esters, benzoates, carbonates, and sulfonates.
Particularly preferred protecting groups include THP.
Bacterial infections and infectious diseases
One of ordinary skill in the art is readily able to determine whether or not a candidate compound treats a bacterial infection or bacterial infectious disease. For example, assays which may conveniently be used to assess the activity offered by a particular compound are described in the examples below.
The term “bacterial infection” pertains to an invasion of body tissues by bacteria, their multiplication and the reaction of body tissues to the bacteria and the toxins that they produce. The term “bacterial infectious disease” pertains to a disease caused by bacteria.
Examples of bacterial infections and infectious bacterial diseases include, but are not limited to, hospital- and community acquired infections due to Gram-positive bacteria such as Staphylococcus aureus, including multi-drug resistant forms such as methicillin-resistant S. aureus (MRSA), and to streptococci, including drug resistant forms such as vancomycin-resistant enterococci (VRE). Other infections that could be treated include those due to Clostridium difficile, Listeria monocytogenes and skin infections due to Gram-positive bacteria.
Preferrably the compounds of the present invention are useful in the treatment of bacterial infections infectious bacterial diseases caused by bacterial infections involving drug resistant bacterial strains. Particularly preferred bacterial strains are Staphylococcus aureus.
Preferably, the treatment is of a bacterial infectious disease or bacterial infection involving MRSA, vancomycin-intermediate S. aureus (VISA), vancomycin-resistant S. aureus (VRSA) and other infections due to drug resistant Gram-positive bacteria.
Methods of Treatment
As described above, the present invention provides the use of a compound of the first aspect of the invention in a method of therapy.
The term “therapeutically effective amount” is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors. A compound may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. Examples of treatments and therapies include, but are not limited to, β-lactam drugs such as the penicillins and cephalosporins, aminoglycosides, fluoroquinolones, oxazolidinones and the streptogramins.
Pharmaceutical compositions according to the present invention, and for use in accordance with the present invention, may comprise, in addition to the active ingredient, i.e. a compound of formula I, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous, or intravenous.
Pharmaceutical compositions for oral administration may be in tablet, capsule, powder or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. A capsule may comprise a solid carrier such a gelatin.
For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
Dosage
It will be appreciated by one of skill in the art that appropriate dosages of the compound can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects. The selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, the severity of the condition, and the species, sex, age, weight, condition, general health, and prior medical history of the patient. The amount of compound and route of administration will ultimately be at the discretion of the physician, veterinarian, or clinician, although generally the dosage will be selected to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.
Administration can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell(s) being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician, veterinarian, or clinician.
In general, a suitable dose of the active compound is in the range of about 100 ng to about 25 mg (more typically about 1 pg to about 10 mg) per kilogram body weight of the subject per day. Where the active compound is a salt, an ester, an amide, a prodrug, or the like, the amount administered is calculated on the basis of the parent compound and so the actual weight to be used is increased proportionately.
In one embodiment, the active compound is administered to a human patient according to the following dosage regime: about 100 mg, 3 times daily.
In one embodiment, the active compound is administered to a human patient according to the following dosage regime: about 150 mg, 2 times daily.
In one embodiment, the active compound is administered to a human patient according to the following dosage regime: about 200 mg, 2 times daily.
For the prevention or treatment of disease, the appropriate dosage of the compound of the invention will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the molecule is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The molecule is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1-20 mg/kg) of molecule is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. An exemplary dosage of compound to be administered to a patient is in the range of about 0.1 to about 10 mg/kg of patient weight. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. An exemplary dosing regimen comprises a course of administering an initial loading dose of about 4 mg/kg, followed by additional doses every week, two weeks, or three weeks of a compound.
Other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
Includes Other Forms
Unless otherwise specified, included in the above are the well known ionic, salt, solvate, and protected forms of these substituents. For example, a reference to carboxylic acid (-COOH) also includes the anionic (carboxylate) form (-COO), a salt or solvate thereof, as well as conventional protected forms. Similarly, a reference to an amino group includes the protonated form (-N+HR1R2), a salt or solvate of the amino group, for example, a hydrochloride salt, as well as conventional protected forms of an amino group. Similarly, a reference to a hydroxyl group also includes the anionic form (-O'), a salt or solvate thereof, as well as conventional protected forms.
Isomers, Salts and Solvates
Certain compounds may exist in one or more particular geometric, optical, enantiomeric, diasteriomeric, epimeric, atropic, stereoisomeric, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans-forms; E- and Z-forms; c-, t-, and r- forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and I-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal-and anticlinal-forms; a- and β-forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and halfchair-forms; and combinations thereof, hereinafter collectively referred to as “isomers” (or “isomeric forms”).
Preferably compounds of the present invention have the following stereochemistry at the C11 position:
Note that, except as discussed below for tautomeric forms, specifically excluded from the term “isomers”, as used herein, are structural (or constitutional) isomers (i.e. isomers which differ in the connections between atoms rather than merely by the position of atoms in space). For example, a reference to a methoxy group, -OCH3, is not to be construed as a reference to its structural isomer, a hydroxymethyl group, -CH2OH. Similarly, a reference to ortho-chlorophenyl is not to be construed as a reference to its structural isomer, meta-chlorophenyl. However, a reference to a class of structures may well include structurally isomeric forms falling within that class (e.g. C1.7 alkyl includes n-propyl and iso-propyl; butyl includes η-, iso-, sec-, and tert-butyl; methoxyphenyl includes ortho-, meta-, and para-methoxyphenyl).
The above exclusion does not pertain to tautomeric forms, for example, keto-, enol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol (illustrated below), imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime, thioketone/enethiol, N-nitroso/hyroxyazo, and nitro/aci-nitro.
Note that specifically included in the term “isomer” are compounds with one or more isotopic substitutions. For example, H may be in any isotopic form, including 1H, 2H (D), and 3H (T); C may be in any isotopic form, including 12C, 13C, and 14C; O may be in any isotopic form, including 160 and 180; and the like.
Unless otherwise specified, a reference to a particular compound includes all such isomeric forms, including (wholly or partially) racemic and other mixtures thereof. Methods for the preparation (e.g. asymmetric synthesis) and separation (e.g. fractional crystallisation and chromatographic means) of such isomeric forms are either known in the art or are readily obtained by adapting the methods taught herein, or known methods, in a known manner.
Unless otherwise specified, a reference to a particular compound also includes ionic, salt, solvate, and protected forms of thereof, for example, as discussed below.
It may be convenient or desirable to prepare, purify, and/or handle a corresponding salt of the active compound, for example, a pharmaceutically-acceptable salt. Examples of pharmaceutically acceptable salts are discussed in Berge, etal., J. Pharm. Sci., 66, 1-19 (1977).
For example, if the compound is anionic, or has a functional group which may be anionic (e.g. -COOH may be -COO'), then a salt may be formed with a suitable cation.
Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such as Na+ and K+, alkaline earth cations such as Ca2+ and Mg2+, and other cations such as Al3+. Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e. NH4+) and substituted ammonium ions (e.g. NH3R+, NH2R2+, NHR3+, NR4+). Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH3)4+.
If the compound is cationic, or has a functional group which may be cationic (e.g. -NH2 may be -NH3+), then a salt may be formed with a suitable anion. Examples of suitable inorganic anions include, but are not limited to, those derived from the following inorganic acids: hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfurous, nitric, nitrous, phosphoric, and phosphorous.
Examples of suitable organic anions include, but are not limited to, those derived from the following organic acids: 2-acetyoxybenzoic, acetic, ascorbic, aspartic, benzoic, camphorsulfonic, cinnamic, citric, edetic, ethanedisulfonic, ethanesulfonic, fumaric, glucheptonic, gluconic, glutamic, glycolic, hydroxymaleic, hydroxynaphthalene carboxylic, isethionic, lactic, lactobionic, lauric, maleic, malic, methanesulfonic, mucic, oleic, oxalic, palmitic, pamoic, pantothenic, phenylacetic, phenylsulfonic, propionic, pyruvic, salicylic, stearic, succinic, sulfanilic, tartaric, toluenesulfonic, and valeric. Examples of suitable polymeric organic anions include, but are not limited to, those derived from the following polymeric acids: tannic acid, carboxymethyl cellulose.
It may be convenient or desirable to prepare, purify, and/or handle a corresponding solvate of the active compound. The term “solvate” is used herein in the conventional sense to refer to a complex of solute (e.g. active compound, salt of active compound) and solvent. If the solvent is water, the solvate may be conveniently referred to as a hydrate, for example, a mono-hydrate, a di-hydrate, a tri-hydrate, etc.
Compounds of formula I include compounds where a nucleophilic solvent (H20, RAOH, RaNH2, RaSH) adds across the imine bond of the PBD moiety, which is illustrated below where the solvent is water or an alcohol (RAOH, where RA is an ether substituent as described above):
These forms can be called the carbinolamine and carbinolamine ether forms of the PBD. The balance of these equilibria depend on the conditions in which the compounds are found, as well as the nature of the moiety itself.
These compounds may be isolated in solid form, for example, by lyophilisation.
General synthetic routes
Compounds of formula I where R10 and R11 together form a double bond may be synthesised from compounds of formula 2:
R’10 is a nitrogen protecting group and R’11 is O-R12, wherein R12 is H or a hydroxyl protecting group. Such techniques are well known in the art, and are described, for example, in Wuts, P.G.M. and Greene, T.W., Protective Groups in Organic Synthesis, 4th Edition, Wiley-lnterscience, 2007. If both nitrogen and hydroxyl protecting groups are present, these are preferably selected to be removable by the same conditions.
If this deprotection is carried out in a solvent of formula HQRq, then R10 and R11 will be H and QRq respectively. Alternatively, these groups may be introduced by adding the compound to a different solvent to that in which the deprotection is carried out.
The conversion of compounds of formula I as discussed above to those having R11 as SOxM may be achieved by the addition of the appropriate bisulphite salt or sulphinate slat, followed by a purification step. Further methods are described in GB 2 053 894, which is herein incorporated by reference.
Compounds of formula 2 can be made by the coupling of compounds of Formula 3 and Formula 4:
under standard amide bond formation conditions, e.g. in the presence of HOBt or DMAP and EDCI.
Compounds of formula 3 can be synthesised from compounds of formula 5:
where R’8 is a Ci.4 alkyl group, e.g. methyl. This deprotection of the carboxyl group may be carried out using standard means, e.g. treatment with base.
Compounds of formula 5 can be synthesised in general following the methods described in WO 00/12506 and WO 2007/039752, which are herein incorporated by reference. In particular, the butanoic acid side chain can be introduced at any stage in the synthesis, usually with appropriate protecting groups in place. For example, the side chain can be formed by coupling a protected or precursor form to a hydroxy group on the bezene ring using e.g. Mitsunobo coupling.
Compounds of formula 4 can be synthesised using the methods disclosed in WO 2005/085177, which are incorporated herein by reference. Reference is also made to the disclosure of WO 2007/039752. DNA Binding
The ability of the compounds to bind to DNA, and in particular oligonucleotides, can be measured using an Ion Pair Reversed-Phase HPLC assay, as described in Rahman, K. M., etal., Journal of the American Chemical Society 2009, 131, 13756 and Narayanaswamy, M., etal., Analytical Biochemistry 2008, 374, 173. The DNA binding affinity can also be evaluated by using a calf-thymus DNA thermal denaturation assay, as described in Wells, G., etal., Journal of Medicinal Chemistry 2006, 49, 5442; Jenkins, T. C., etal., Journal of Medicinal Chemistry 1994, 37, 4529; and Gregson, S. J., etal., Journal of Medicinal Chemistry 2001, 44, 737.
Further preferences C2
It may be preferred in any of the embodiments that the C2 carbon is a sp2 centre, so that when R2 is selected from any of the following groups: -H, -OH, -CN, -R, -OR, halo, -0-S02-R, -C02R and -COR there is a double bond between C2 and C3.
When R2 is selected from any of the following groups: =0, =CH2, =CHR, =CHRR’ there cannot be a double bond between C2 and C3. R2 R2 is selected from -H, -OH, =0, =CH2, -CN, -R, OR, halo, dihalo, =CHR, =CHRR’, -O-S02-R, C02R and COR.
In some embodiments, R2 may be selected from -H, -OH, =0, =CH2, -CN, -R, -OR, =CHR, =CRR\ -0-S02-R, -C02R and -COR.
In some embodiments, R2 may be selected from -H, =CH2, -R, =CHR, and =CRR’.
In one embodiment, R2 is H.
In one embodiment, R2 is =0.
In one embodiment, R2 is =CH2.
In one embodiment, R2 is =CHR. Within the PBD compound, the group =CHR may have either configuration shown below:
In one embodiment, the configuration is configuration (C1).
In one embodiment, R2 is =CRR’.
In one embodiment, R2 is =CF2.
In one embodiment, R2 is R.
In one embodiment, R2is optionally substituted C5-20 aryl.
When R2 is optionally substituted C5.2o aryl, it may preferably be optionally substituted C5.7 aryl or C8.io aryl. R2 may further preferably be optionally substituted phenyl, optionally substituted napthyl, optionally substituted pyridyl, optionally substituted quinolinyl or isoquinolinyl. Of these groups, optionally susbtitued phenyl is most preferred.
When R2 is optionally substituted C5.20 aryl, it may preferably bear one to three substituent groups, with 1 and 2 being more preferred, and singly substituted groups being most preferred. The substituents may be any position.
Where R2 is a C5.7 aryl group, a single substituent is preferably on a ring atom that is not adjacent the bond to the remainder of the compound, i.e. it is preferably β or γ to the bond to the remainder of the compound. Therefore, where the C5.7 aryl group is phenyl, the substituent is preferably in the meta- or para- positions, and more preferably is in the para- position.
In one embodiment, R2 is selected from:
where the asterisk indicates the point of attachment.
Where R2 is a C8-io aryl group, for example quinolinyl or isoquinolinyl, it may bear any number of substituents at any position of the quinoline or isoquinoline rings. In some embodiments, it bears one, two or three substituents, and these may be on either the proximal and distal rings or both (if more than one substituent).
When R2 is optionally substituted C5.2o aryl, the substituents may be selected from: halo, hydroxyl, ether, formyl, acyl, carboxy, ester, acyloxy, amino, amido, acylamido, aminocarbonyloxy, ureido, nitro, cyano and thioether.
When R2 is optionally substituted C5.2o aryl, the substituents may be selected from the group consisting of R, OR, SR, NRR’, N02, halo, C02R, COR, CONH2, CONHR, and CONRR’.
If a substituent on R2 is halo, it is preferably F or Cl, more preferably Cl.
If a substituent on R2 is ether, it may in some embodiments be an alkoxy group, for example, a Ci_7 alkoxy group (e.g. methoxy, ethoxy) or it may in some embodiments be a C5.7 aryloxy group (e.g phenoxy, pyridyloxy, furanyloxy).
If a substituent on R2 is C1.7 alkyl, it may preferably be a C1.4 alkyl group (e.g. methyl, ethyl, propyl, butyl).
If a substituent on R2 is C3-7 heterocyclyl, it may in some embodiments be C6 nitrogen containing heterocyclyl group, e.g. morpholino, thiomorpholino, piperidinyl, piperazinyl. These groups may be bound to the rest of the PBD moiety via the nitrogen atom. These groups may be further substituted, for example, by C1.4 alkyl groups.
If a substituent on R2 is bis-oxy-C^ alkylene, this is preferably bis-oxy-methylene or bis-oxy-ethylene.
Particularly preferred substituents for R2 include methoxy, ethoxy, fluoro, chloro, cyano, bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thienyl.
Particularly preferred substituted R2 groups include, but are not limited to, 4-methoxy-phenyl, 3-methoxyphenyl, 4-ethoxy-phenyl, 3-ethoxy-phenyl, 4-fluoro-phenyl, 4-chloro-phenyl, 3,4-bisoxymethylene-phenyl, 4-methylthienyl, 4-cyanophenyl, 4-phenoxyphenyl, quinolin-3-yl and quinolin-6-yl, isoquinolin-3-yl and isoquinolin-6-yl, 2-thienyl, 2-furanyl, methoxynaphthyl, and naphthyl.
In one embodiment, R2is optionally substituted Cm2 alkyl.
When R2is optionally substituted Cm2 alkyl, it may be selected from: (a) C1-5 saturated aliphatic alkyl; (b) C3-6 saturated cycloalkyl; (c)
, wherein each of R21, R22 and R23 are independently selected from H, C1. 3 saturated alkyl, C2.3 alkenyl, C2.3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R12 group is no more than 5; (d)
, wherein one of R25a and R25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo methyl, methoxy; pyridyl; and thiophenyl; and (e)
where R24 is selected from: H; C1.3 saturated alkyl; C2.3 alkenyl; C2.3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo methyl, methoxy; pyridyl; and thiophenyl.
When R12 is C1.5 saturated aliphatic alkyl, it may be methyl, ethyl, propyl, butyl or pentyl. In some embodiments, it may be methyl, ethyl or propyl (n-pentyl or isopropyl). In some of these embodiments, it may be methyl. In other embodiments, it may be butyl or pentyl, which may be linear or branched.
When R12 is C3.6 saturated cycloalkyl, it may be cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. In some embodiments, it may be cyclopropyl.
When R12 is
, each of R21, R22 and R23 are independently selected from H, C1-3 saturated alkyl, C2.3 alkenyl, C2.3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R12 group is no more than 5. In some embodiments, the total number of carbon atoms in the R12 group is no more than 4 or no more than 3.
In some embodiments, one of R21, R22 and R23 is H, with the other two groups being selected from H, C1.3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl.
In other embodiments, two of R21, R22 and R23 are H, with the other group being selected from H, Ci.3 saturated alkyl, C2.3 alkenyl, C2.3 alkynyl and cyclopropyl.
In some embodiments, the groups that are not H are selected from methyl and ethyl. In some of these embodiments, the groups that are not H are methyl.
In some embodiments, R21 is H.
In some embodiments, R22 is H.
In some embodiments, R23 is H.
In some embodiments, R21 and R22 are H.
In some embodiments, R21 and R23 are H.
In some embodiments, R22 and R23 are H.
When R12 is
, one of R25a and R25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl. In some embodiments, the group which is not H is optionally substituted phenyl. If the phenyl optional substituent is halo, it is preferably fluoro. In some embodiment, the phenyl group is unsubstituted.
When R12 is
R24 is selected from: H; Ci.3 saturated alkyl; C2.3 alkenyl; C2.3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo methyl, methoxy; pyridyl; and thiophenyl. If the phenyl optional substituent is halo, it is preferably fluoro. In some embodiment, the phenyl group is unsubstituted.
In some embodiments, R24 is selected from H, methyl, ethyl, ethenyl and ethynyl. In some of these embodiments, R24 is selected from H and methyl.
In one embodiment, R2 is halo or dihalo. In one embodiment, R2 is -F or -F2, which substituents are illustrated below as (C3) and (C4) respectively:
R2 may preferably selected from =CH2, =CH-R, where R is more preferably an optionally substituted Ci_4 alkyl group, and -R, where R is more preferably an optionally substituted C5-20 aryl group. Particularly preferred groups for R2 include =CH2, =CH-Me, and an optionally substituted phenyl group. R7 R7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR’, nitro, Me3Sn and halo; R7 may preferably be selected from H, OR, SH, SR, NH2, NHR, NRR’, and halo. R7 may more preferably be selected from H and OR.
In some embodiments, R7 is OR, and more particularly OR7A, where R7A is independently optionally substituted C1.7 alkyl. R7A may be selected from optionally substituted saturated C1.7 alkyl and optionally substituted C2.4 alkenyl. R7A may preferably be selected from Me, CH2Ph and allyl. R10/R11 R10 and R11 either together form a double bond, or are selected from H and QR° respectively, where Q is selected from O, S and NH and R° is H or C1.7 alkyl or H and SOxM, where x is 2 or 3, and M is a monovalent pharmaceutically acceptable cation;
In some embodiments, R10 and R11 form a double bond together.
In some embodiments, R10 is H and R11 is OR0. In these embodiments, R° may preferably be selected from H or Me.
In some embodiments, R10 is H and R11 is SOxM. x may preferably be 3, and M may preferably be Na+. R1 R1 is C1.4 alkyl. R1 may preferably be C1.2 alkyl, and more preferably methyl.
A A is selected from A1, A2, A3, A4 or A5. X1 and Ϋ
For each of A1, A2, A3, A4 and A5, X1 and Y1 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and 0, respectively.
Preferably X1 and Y1 are selected from: CH and NMe; N and NMe; CH and S; and CH and 0, respectively. Most preferably, X1 and Y1 are selected from N and NMe and CH and NMe, respectively.
F F is selected from a single bond and -(E-F1)m-.
When F is -(E-F1)m-, each E is independently selected from a single bond, and -C(=0)-NH-; each F1 is independently a C3.2o heteroarylene group; and m is 1, 2 or 3.
Preferably, F1 is a C5-e heteroarylene group, more prefereably a C5 heteroarylene group. More preferably, F1 is represented by structure F11:
(F11) wherein X3 and Y3 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and O, respectively. Preferably X3 and Y3 are selected from: CH and O; and CH and NMe, respectively.
Thus, preferred structures of F11 are:
Each E is independently selected from a single bond and -C(=0)-NH-. In some embodiments, each E is -C(=0)-NH-. m is 1, 2 or 3. Preferably m is 1. In other embodiments, m is 3.
In some embodiments F is a single bond.
G G is selected from hydrogen, C1.4alkyl, -C(=0)-0-Ci.4alkyl, -(CH2)n-C3-2o heterocycloalkyl, and -0-(CH2) n_C3-2o heterocycloalkyl group; and each n is 0-4.
Preferably, the C3.20 heterocycloalkyl group is a morphiline group. Preferably, G is selected from the group consisting of-H, -Me, -C(=0)-0-Me, -(CH2)n-N(CH2CH2OCH2CH2), and -0-(CH2)n-N(CH2CH20CH2CH2). n is 0-4. Thus, n may be 0, 1,2,3 or 4.
Preferred combinations of F and G
In some embodiments, G and at least one of the (-E-F1)- units of F are provided by the structures FG1 and FG2:
FG1 and FG2 may be combined with other -(E-F1)- units of F if m is 2 or 3. A1 A1 is represented by the structure:
Z1 is O or S. Preferably X1 and Y1 are selected from CH and NMe, and N and NMe.
Thus preferred structures of A1 are A11, A12, A13 and A14:
A2 A2 is represented by the structure:
Preferably X1 and Y1 are selected from CH and NMe, and N and NMe. Thus, preferred strucutres of A2 are A21, A22, A23 and A24:
Preferably -F-G is arranged in the para-position of the phenylene or pyridinyl group of A2.
When A2 is represented by structures A21 or A23, A may not be A2’:
where X1 and Y1 of A2’ are selected from: CH and NMe and N and NMe, respectively; B is either a single bond or:
where X and Y of (B1) are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; and R1 is C1.4 alkyl. A3 A3 is represented by the structure:
Preferably, X1 and Y1 are selected from CH and NMe, N and NMe, and CH and O. Preferably X2 and Y2 are selected from CH and NMe, and N and NMe.
Thus preferred structures of A3 are A31, A32, A33, A34, A35 and A36:
Preferably the PBD portion of the compound is arranged in the para-position of the phenylene group of A3.
When A3 is represented by structures A31 or A32, A may not be A3’:
where X2 and Y2 of A3’ are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; B is either a single bond or:
where X and Y of B1 are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; and R1 is C1.4 alkyl. A4 A4 is represented by the structure:
Preferably, X1 and Y1 are CH and 0 and Z1 is NMe. Thus a preferred structure of A4 is A41:
A5 is represented by the structure
Preferably X1 and Y1 are selected from CH and NMe, and CH and O. So preferred structures of A5 are given as A51, A52, A53 and A54:
When Z2 of A5 is CH, F is preferebaly a single bond. Preferred compounds of the present invention are as follows:
Compounds 26, 27, 28, 35, 54, and 56 are preferred. Compounds 26, 27, and 35 are more preferred, and compound 35 is particularly preferred.
Examples
General methods
Optical rotations were measured on an ADP 220 polarimeter (Bellingham Stanley Ltd) and concentrations (c) are given in g/100mL. Melting points were measured using a digital melting point apparatus (Electrothermal). IR spectra were recorded on a Perkin-Elmer Spectrum 1000 FT IR Spectrometer. 1H and 13C NMR spectra were acquired at 300 K using a Bruker Advance NMR spectrometer at 400 and 100 MHz, respectively. Chemical shifts are reported relative to TMS (δ = 0.0 ppm), and signals are designated as s (singlet), d (doublet), t (triplet), dt (double triplet), dd (doublet of doublets), ddd (double doublet of doublets) or m (multiplet), with coupling constants given in Hertz (Hz). A pro-PBD numbering system is used for carbon and proton assignments for synthetic intermediates (/.e., based on the final tricyclic PBD ring system). Mass spectrometry data were collected using a Waters Micromass ZQ instrument coupled to a Waters 2695 HPLC with a Waters 2996 PDA. Waters Micromass ZQ parameters used were: Capillary (kV), 3.38; Cone (V), 35; Extractor (V), 3.0; Source temperature (°C), 100; Desolvation Temperature (°C), 200; Cone flow rate (L/h), 50; De-solvation flow rate (L/h), 250. High-resolution mass spectrometry data were recorded on a Waters Micromass QTOF Global in positive W-mode using metal-coated borosilicate glass tips to introduce the samples into the instrument. Thin Layer Chromatography (TLC) was performed on silica gel aluminum plates (Merck 60, F254), and flash chromatography utilized silica gel (Merck 60, 230-400 mesh ASTM). Parallel reactions were carried out using a Radleys™ Green House Synthesizer and parallel purifications were carried out using an 1ST Vacmaster™. For reactions carried out in parallel, solvents were evaporated using a Genevac VC 2000D (Genevac Technologies, UK). Purified compounds were freeze dried using a Heto-Lyolab 3000 freeze drier. Hydrogenation reactions were carried using UHP-60H hydrogen generator attached to a Parr hydrogenation apparatus. Synthetic building blocks were purchased from Maybridge Chemicals (UK), Bachem Chemicals (USA) and Sigma-Aldrich (UK). Reagents and solvents were purchased from Sigma-Aldrich (UK).
Synthesis of Intermediates
A solution of N-methyl pyrrole (1)(113.06 g, 1.39 mol, 1.0 eq) in dry ether (350 mL) was added drop wise over a period of 1 hour and 10 minutes to a stirred solution of trichloroacetyl chloride (254 g, 1.39 mol, 1.0 eq) in dry ether (350 mL) in a 2 L, 3 necked flask. HCI gas produced in the reaction was removed by flushing with nitrogen. The reaction mixture was allowed to stir for 1.5 hours and progress of the reaction was monitored regularly by TLC and LCMS. After 1.5 hours the reaction was quenched using 1 M K2C03 solution. The reaction mixture was extracted with ethyl acetate (3 x) and the organic layers were combined and concentrated in vacuo. The crystalline residue was washed with n-hexane and finally dried under vacuum. Yield - 281.18 g, 79.5%, NMR compared with literature IR, (FTIR, vmax /cm'1) 3299, 3121, 3008, 2954, 1789, 1674, 1521, 1406, 1206, 1100, 980, 881, 757; 1H-NMR (CDCI3, 400 MHz) δ 7.42 (1H, dd, J = 4.4, 1.6 Hz), 6.97 (1H, t, J = 1.6 Hz), 6.22 (1H, dd, J = 4.4, 2.4 Hz) 3.97 (3H, s); 13C NMR (CDCI3, 400 MHz): δ 133.6, 124.0, 122.4, 108.9, 38.5. (ii) 1 -(4-bromo-1 -methyl-1 H-pyrrol-2-yl)-2,2,2-trichloroethanone (3) NBS (N-Bromosuccinimide, 2.36 g, 13.24 mmol, 1.0 equiv.) was added to a stirred solution of 2-(trichloroacetyl)-1-methylpyrrole (2) (3 g, 13.24 mmol, 1.0 equiv.) in anhydrous THF (35 mL) at -10°C. The reaction mixture was kept at -10°C for 2 hours and then left to reach room temperature (ca. 4 hours). Excess THF was evaporated in vacuum and the solid was re-dissolved in a mixture of EtOAc/n-hexane (1:9). The resulting mixture was filtered through a plug of silica, and the filtrate was evaporated in vacuo. The resulting solid was recrystallised from n-hexane to give 3 (3.55 g, 88%). IR (FTIR, vmax/cm"1): 3148, 2956, 1669 (0=0),1458, , 1215, 1189, 1062, 923, 842, 823, 748, 678; 1H-NMR (CDCI3, 400 MHz) δ 7.46 (1H, d, J = 2.0 Hz), 6.95 (1H, d, J = 1.6 Hz) 3.95 (3H, s); 13C NMR (CDCI3, 100 MHz): δ 172.4, 132.8, 124.6, 132.2, 96.1, 38.7; EIMS m/z (+EI) calc, for C7H5BrCI3NO (M)+ 305.38, LCMS analysis found 306.86 (M+H)+ (Hi) Methyl 4-bromo-1 -methyl-1 H-pyrrole-2-carboxylate (4)
To a stirred solution of 1-(4-bromo-1-methyl-1H-pyrrol-2-yl)-2,2,2-trichloro-ethanone (3)(3.28 g, 10.74 mmol, 1 eq.) in dry MeOH (30 ml_), a solution of sodium methoxide (0.5 mL) was added by a syringe. The sodium methoxide solution was prepared from NaH 60% in mineral oil (43 mg, 1.07 mmol, 0.1 eq.), which was previously washed with n-hexane. The solution was heated to reflux over a period of 30 minutes, when the TLC analysis showed complete consumption of the starting material. A few drops of concentrated H2S04 were added to the solution to neutralise the base (pH 2). The excess MeOH was evaporated in vacuum and the resulting oil was redissolved in EtOAc (50 mL) and washed with water (40 mL). The aqueous layer was extracted with EtOAc (3 x 40 mL), and the organic phases were combined, dried (MgS04), filtered and concentrated in vacuum to afford the product as a pale white solid. (2.28 g, 97%). IR (FTIR, Vmax/cm"1): 3138, 2948, 1692, 1472, 1334, 1245, 1115, 1082, 921, 823, 753; 1H-NMR (400 MHz, CDCI3): δ 6.89 (d, 1H, J = 2.0 Hz), 6.76 (d, 1H, J = 2.0 Hz), 3.89 (s, 3H), 3.81 (s, 3H); 13C-NMR (100 MHz, CDCI3): δ 160.8, 128.7, 122.9, 119.2, 95.1, 51.2, 36.9; EIMS m/z (+EI) calc, for C7H8BrN02 (M)+ 218.05 found 219.26 (M+H)+ (iv) Methyl 4-(4-(tert-butoxycarbonylamino)phenyl)-1 -methyl-1 H-pyrrole-2-carboxylate (5) A catalytic amount of tetrakis(triphenylphosphine)palladium, Pd(PPh3)4 (0.477g, 0.413, 0.06 eq) was added to a solution of 4 (1.5 g, 6.88 mmol, 1 eq) and (4-((tert-butoxycarbonyl)amino)phenyl)boronic acid (1.57 g, 6.88 mmol, 1.20 eq) in a 9:3:1 combination (13.5 ml) of EtOH, toluene and water in the presence of K2C03 (2.856 g, 3 eq.) in a 10-20 mL microwave vial containing a magnetic stirrer. The reaction vessel was flushed with nitrogen during each addition. The reaction mixture was sealed in an inert N2 atmosphere and heated with microwave radiation in an EMRYS™ Optimizer Microwave Station (Personal Chemistry) at 100°C for 12 minutes. After LCMS and TLC analysis revealed completion of the reaction, the cooled reaction mixture was diluted with water (50 mL), extracted with EtOAc (3 x 40 mL), the filtrates combined, dried over MgS04 and concentrated under vacuum. The resulting oil was subjected to flash chromatography (n-hexane/EtOAc 9:1) to give 5 (Yield - 2.2 g, 97%). IR (FTIR, vmax /cm" 1): 3353, 2975, 1696, 1521, 1441, 1366, 1264, 1235, 1209, 1058, 822, 799, 657; 1H-NMR (400 MHz, CDCI3): δ 7.40 (d, 2H, J = 8.8 Hz), 7.33 (d, 2 H, J = 8.8 Hz), 7.16 (d, 1H, J = 2.0 Hz,), 7.02 (d, 1H, J = 2.0), 6.45 (brs, 1H), 3.95 (s, 3H), 3.83 (s, 3H), 1.52 (s, 9H); 13C-NMR (100 MHz, CDCIs): δ 161.7, 152.8, 136.5, 129.5, 125.9, 125.6, 123.7, 123.0, 119.0, 114.6, 80.5, 51.1, 36.9, 28.4; EIMS m/z (+EI) calc, for Ci^NsC^ (M)+ 330.38 found 330.46 (M+H)+
A 0.5 M solution of NaOH (2.0 eq) was added to a solution of 5 (1.0 g, 3.027 mmol) in dioxane (40 mL). The reaction mixture was allowed to stir at room temperature for 6 hours at which point TLC revealed completion of reaction. Excess 1,4-dioxane was evaporated under vacuum and the residue was diluted with water. The resulting solution was acidified with 0.5 M HCI. The product was extracted from water with 2 x ethyl acetate (100 mL x 2) and the organic layers were combined, washed with brine, dried over MgS04 and concentrated in vacuo. The product was purified using flash chromatography (ethyl acetate/n-hexane 2:8). Yield - 0.92 g, 96.8%. IR (FTIR, vmax /cm’1): 3371, 2979, 1698, 1671, 1522, 1445, 1367, 1285, 1161, 1112, 1047, 823, 803, 762, 714, 631; 1H-NMR (400 MHz, CDCI3): δ 8.33 (1H, s), 7.55 (d, 2H, J = 8.8 Hz), 7.50 (d, 2 H, J = 8.8 Hz), 7.36 (d, 1H, J = 2.0 Hz,), 7.22 (d, 1H, J = 2.0), 3.97 (s, 3H), 1.50 (s, 9H); 13C-NMR (100 MHz, CDCI3): δ 162.3, 153.7, 138.6, 123.0, 127.1, 126.0, 124.4, 124.0, 119.5, 115.1, 79.9, 36.9, 28.6; EIMS m/z (+EI) calc, for C17H20N2O4 (M)+ 316.35 found 315.16 (M+H)+ (C)
(i) Methyl 4-(4-aminophenyl)-1 -methyl-1 H-pyrrole-2-carboxylate (7) 5 (1 g, 3.027 mmol) was dissolved in a small volume of MeOH and 4M HCI in dioxane (15 ml_) was added slowly to the stirring solution. The reaction mixture was stirred for 6 hours at which point TLC showed completion of reaction. Excess solvent was evaporated under vacuum to obtain a brown coloured solid 7. The solid product was subjected to flash chromatography (n-hexane/EtOAc 9:1) to give pure 7 (065 g, 94.2%). IR (FTIR, vmax /cm"1): 3366, 2987,1688, 1629, 1566, 1422, 1372, 1262, 1181, 1103, 1067, 951, 821, 784, 756; 1H-NMR (400 MHz, CDCI3): δ 7.28 (2H, d, J = 8.4 Hz), 7.11 (1H, d, J = 2.0 Hz), 6.96 (1H, d, J = 2.0 Hz), 6.68 (d, 2 H, J = 8.0 Hz), 3.94 (s, 3H), 3.83 (s, 3H); 13C-NMR (100 MHz, CDCI3): δ 161.7, 144.7, 126.2, 125.4,125.2, 115.5, 114.4, 51.0, 36.8; EIMS m/z (+EI) calc, for Ci3Hi4N202 (M)+230.26 found 231.1 (M+H)+ (d)
(i)
Methyl 4-(4-(4-aminophenyl)-1 -methyl-1 H-pyrrole-2-carboxamido)-1 -methyl-1 H-pyrrole-2-carboxylate (9)
Two eq of EDCI and 2.5 eq of DMAP were added to a stirred solution of 6 (0.45 gm, 1.2 eq) in DMF (8 ml_) and the mixture was allowed to stir for 30 minutes after which commercially available methyl 4-amino-1 -methyl-1 H-pyrrole-2-carboxylate (0.18 g, 1.18 mmol, 1.0 eq) was added. The reaction mixture was allowed to stir for a further 6 hour at which point TLC showed completion of reaction. The reaction was quenched by pouring it onto a mixture of ice/water mixture and the resulting mixture was extracted with ethyl acetate (3 x 150 ml_). The combined extracts were sequentially washed with citric acid (100 ml_), saturated aqueous NaHC03 (100 ml_), water (100 ml_), brine (100 ml_) and finally dried over MgS04. Excess ethyl acetate was evaporated by rotary evaporator under reduced pressure and the crude product 9a (0.58 gm, yield 90.6%) was used for the boc deprotection step to afford 9 without further purification. For boc deprotection, 0.29 gm of 9a was dissolved in a small volume of MeOH and 4M HCI in dioxane (15 ml_) was added slowly to the stirring solution. The reaction mixture was stirred for 6 hours at which point TLC showed completion of reaction. Excess solvent was evaporated under vacuum to obtain a brown coloured solid (9). The solid product was subjected to flash chromatography (n-hexane/EtOAc 9:1) to give pure 9. Yield 0.21 gm, 95%. (ii) Methyl 4-(4-(4-(4-aminophenyl)-1 -methyl-1 H-pyrrole-2-carboxamido)-1 -methyl- 1H-pyrrole-2-carboxamido)-1 -methyl-1 H-pyrrole-2-carboxylate (10)
Lithium hydroxide (68 mg, 1.65 mmol, 3 eq) was added to 9a (0.25 g, 0.55 mmol) in aqueous dioxane (8 ml dioxane, 4 ml water) at room temperature. The reaction mixture was stirred for 3 hours at which point TLC showed completion of reaction. Dioxane was evaporated under high vacuum and the residue was diluted with water. The resulting solution was acidified with 1 M citric acid followed by extraction with ethyl acetate (2 x 50 mL). The organic layer combined and washed with brine (50 mL), dried over MgS04 and finally concentrated using a rotary evaporator under reduced pressure to obtain the hydrolysed acid from of 9a as a white solid (yield 0.23 g, 91.6%). To a stirring solution of the white solid (0.23 gm, 0.52 nmol) in DMF, 2.0 equivalent of EDCI and 2.5 Equivalent of DMAP was added. After stirring the mixture for 20 minutes, commercially available methyl 4-amino-1-methyl-1H-pyrrole-2-carboxylate (80.1 mg, 0.52 mmol, 1.0 eq) was added. The reaction mixture was allowed to stir for a further 3 hour at which point TLC showed completion of reaction. The reaction was quenched by pouring it onto a mixture of ice/water mixture and the resulting mixture was extracted with ethyl acetate (3 x 50 mL). The combined extracts were sequentially washed with saturated aqueous NaHC03 (50 mL) and brine (50 mL) and finally dried over MgS04. Excess ethyl acetate was evaporated by rotary evaporator under reduced pressure and the crude product was used for the boc deprotection step to afford 10. For boc deprotection, the crude intermediate was dissolved in a small volume of MeOH and 4M HCI in dioxane (5 mL) was added slowly to the stirring solution. The reaction mixture was stirred for 3 hours at which point TLC showed completion of reaction. Excess solvent was evaporated under vacuum to obtain a brown coloured solid (10). The solid product was subjected to flash chromatography (n-hexane/EtOAc 8:2) to give pure 10. Yield 0.20 gm, 83% over 2 steps. 1H-NMR (DMSO, 400 MHz): δ 9.72 (1H, s), 8.09 (1H, t,J = 5.6 Hz), 7.71 (2H, d, J = 8.8 Hz), 7.49 (2H, d, J = 8.8 Hz), 7.40 (1H, d, J = 2.0 Hz), 7.27 (1H, d, J = 2.0), 7.19 (1H, d, J = 2.0), 7.03 (1H, dd, J = 4.0, 1.6 Hz), 7.00 (1H, t, J= 2.0 Hz), 6.84 (1H, d, J = 2.0 Hz), 6.10 (1H, m), 3.89 (3H, s). m/z (+EI) calc, for C25H26N6O4 (M)+ 474.51 found 475.35 ([M+H]+
Methyl 4-(4-(4-aminophenyl)-1 -methyl-1 H-pyrrole-2-carboxamido)-1 -methyl-1 /-/-imidazole-2-carboxylate (11) 0.3gm of boc protected 6 (0.94 mmol, 1.2 eq) was dissolved in DMF (5 ml_) to which 2.0 eq of EDCI and 2.5 eq of DMAP were added. The mixture was allowed to stir for 30 minutes after which commercially available methyl 4-amino-1 -methyl-1 H-imidazole-2-carboxylate (0.121 g, 0.79 mmol, 1.0 eq) was added. The reaction mixture was allowed to stir for a further 6 hour at which point TLC showed completion of reaction. The reaction was quenched by pouring it onto a mixture of ice/water mixture and the resulting mixture was extracted with ethyl acetate (3 x 150 ml_). The combined extracts were sequentially washed saturated aqueous NaHC03 (50 ml_), water (50 ml_), brine (50 mL) and finally dried over MgS04. Excess ethyl acetate was evaporated by rotary evaporator under reduced pressure and the crude product (0.48 gm) was used for the boc deprotection step to afford 11. For boc deprotection, the crude intermediate was dissolved in a small volume of MeOH and 4M HCI in dioxane (5 mL) was added slowly to the stirring solution. The reaction mixture was stirred for 2 hours at which point TLC showed completion of reaction. Excess solvent was evaporated under vacuum to obtain a brown coloured solid (11). The solid product was subjected to flash chromatography (n-hexane/EtOAc 9:1) to give pure 11. Yield 0.35 gm, 81% over two steps. 1H-NMR (DMSO, 400 MHz): 9.75 (1H, s), 8.03 (1H, s), 7.71 (2H, d, J = 8.8 Hz, 7.53 (1H, s), 7.52 (1H, s), 7.48 (2H, d, J = 8.8 Hz), 7.42 (1H, s), 7.19 (1H, d, J = 2.0), 3.94 (3H, s), 3.91 (3H, s), 3.89 (3H, s). m/z (+EI) calc, for C18H19N5O3 (M)+ 353.38 found 354.42 ([M+H]+ (f) 4-(10-(allyloxycarbonyl)-7-methoxy-5-oxo-11-(tetrahydro-2H-pyran-2-yloxy)- 2,3,5,10,11,11 a-hexahydro-1 H- pyrrolo[2,1-c][1,4]benzodiazepine-8-yloxy)butanoic acid (13)
A 0.5 M solution of NaOH (made from 1.4135 g of NaOH) was added to a solution of 12 (Compound 18, WO 2007/039752) in dioxane at room temperature. The reaction mixture was allowed to stir for 4 hours at which point TLC showed completion of the reaction. Dioxane was evaporated under high vacuum and the residue was diluted with water. The resulting solution was acidified with 1 M citric acid followed by extraction with ethyl acetate (2 x 100 ml_). The combied organic layers were washed with brine (100 ml_), dried over MgS04 and finally concentrated using a rotary evaporator under reduced pressure. Yield - 8.7 g, (94%), 1H-NMR (400 MHz, CDCI3): δ 7.2 (2H, s), 6.90 (1H, s), 6.58 (1H, s), 5.85 (2H, d, J = 9.2 Hz), 5.73 (2H, d, J = 9.2 Hz), 5.03-5.13 (m, 6H), 4.68-4.35 (m, 4H), 4.09-4.01 (m, 4H), 3.91-3.82 (m, 8H), 3.69-3.46 (m, 8H), 2.60-2.55 (m, 4H), 2.18-2.00 (m, 10H), 1.76-1.55 (m, 4H), 1.53-1.43 (m, 8H); 13C-NMR (100 MHz, CDCI3): δ 177.6, 167.6, 149.8, 132.1, 131.9, 126.7, 117.3, 114.9, 110.8, 100.7, 96.0, 91.7, 88.5, 67.9, 66.6, 63.6, 60.1, 56.1, 46.5, 31.1, 30.3, 28.8, 25.2, 24.1, 23.2, 20.0; EIMS m/z (+EI) calc, for C26H34N209 (M)+ 518.56 found 519.26 (M+H)+
Other Q1 and Q2 amine building blocks are commercially available or synthesized analogously.
Example 1 (Comparative compounds)
General Experimental procedure for coupling different building blocks to Alloc-THP protected PBD acid to different building blocks. Example given here is for 15a. Similar procedures were followed to obtain other C8-linked Alloc-THP protected PBD-hvbrids. (11aS)-allyl 7-methoxy-8-(4-(4-(5-(methoxycarbonyl)-1-methyl- 1H-pyrrol-3-yl)phenylamino)-4-oxobutoxy)-5-oxo-11-(tetrahydro-2H-pyran-2-yloxy)-2,3,11,11a-hexahydro-1 H-pyrrolo[2,1 -c][1,4]benzodiazepine-10(5H)-carboxylate (15a) A solution of Alloc-THP protected PBD acid 13 (3.72 g, 7.16 mmol, 1.2 equivalent) was dissolved in DMF. EDCI (2.49 g, 13.02 mmol, 2.0 eq) and DMAP (1.989 g, 16.28 mmol, 2.5 eq) were added to the stirred solution of 13 at room temperature and the mixture was allowed to stir for 30 minutes after which the MPB-ester 7 (1.5 g, 6.514 mmol, 1.0 eq) was added. The reaction mixture was allowed to stir for a further 2 hour at which point TLC showed completion of reaction. The reaction was quenched by pouring it onto a mixture of ice/water mixture and the resulting mixture was extracted with ethyl acetate (3 x 150 ml_). The combined extracts were sequentially washed with citric acid (200 ml_), saturated aqueous NaHC03 (250 ml_), water (250 ml_), brine (250 ml_) and finally dried over MgS04. Excess ethyl acetate was evaporated by rotary evaporator under reduced pressure and the crude product was purified by silica gel flash chromatography (MeOH: CHCI3,20:80) to give a white foamy solid, 15a. Yield -4.05 g, 85.5%. (FTIR, vmax/crTf1): 2949, 2362, 1704, 1600, 1514, 1436, 1372, 1269, 1203, 1107, 1021, 964, 765. (1H NMR, 400 MHz, CDCI3): δ 7.82 (1H, s), 7.48 (2H, m), 7.41 (1H, d, J= 2.0 Hz), 7.40 (1H, d, J= 2.4 Hz), 7.23 (2H, d, J= 8.4 Hz), 7.17 (1H, d, J= 2.0 Hz), 7.04 (1H, d, J= 2.0 Hz), 5.93-5.65 (2H, m), 5.09-5.4.97 (m, 4H), 4.68-4.32 (m, 4H), 4.15-4.10 (m, 4H), 3.94-3.82 (m, 12H), 3.68 (m, 2H), 3.59-3.49 (m, 6H), 2.60-2.57 (m, 3H), 2.15-2.00 (m, 8H), 1.88-1.80 (m, 2H), 1.79- 1.70 (6H), 1.60-1.44 (m, 12H); (13C NMR, 100 MHz, CDCIs): 177.1, 170.5, 167.3, 161,6, 149.1, 136.3, 132.1, 131.9, 130.4, 128.9, 127.1, 125.9, 125.4, 123.5, 123.1, 120.3, 117.3, 114.6, 110.8, 91.5, 88.6, 68.2, 66.5, 64.3, 63.6, 60.3, 56.0, 51.1, 46.4, 36.8, 31.1, 30.9, 29.1, 25.1, 24.6, 23.2, 21.0, 20.1; m/z (+EI) calc, for C39H46N4O10 (M)+ 730.80 found 731.67 ([M+H]+
General Experimental Procedure to obtain the final PBD imine by deprotectina the C8-linked Alloc-THP protected PBD hybrid. Example given here is for 16.
Palladium tetrakis[triphenylphosphine] (5.60 mg, 4.8 μΜ, 0.05 equiv) was added to a solution of of Alloc-THP-PBD conjugate 15a (70 mg, 0.097 mmol), pyrrolidine (8.36 mg, 0.117 mmol, 1.2 eq) and triphenylphosphine (8.62 mg, 0.25 equiv) in DCM (5 ml_). The reaction mixture was stirred at room temperature for 2 hours at which point TLC showed completion of reaction. Excess DCM was removed by rotary evaporation under reduced pressure and the resulting residue dried in vacuo to remove pyrrolidone. The product was purified by column chromatography (eluted with n-hexane 65%%, EtOAc 35%) to give the product as a yellowish solid, 3.37 (40 mg, 77%). [a]22 7D + 165° (c = 0.046, CHCI3); IR (FTIR, vmax/crrf1): 3297, 2944, 2358, 1701, 1598, 1567, 1508, 1442, 1374, 1264, 1212, 1181, 1106, 1072, 824, 730; 1H-NMR (500 MHz, CDCI3): δ 7.68 (1H, s), 7.65 (1H, d, J = 4.5 Hz, H-11), 7.52 (1H, s, H-6), 7.46 (2H, dd, J = 8.4, 2.0 Hz, 2Ar-H), 7.40 (2H, dd, J = 8.4, 2.0 Hz, 2Ar-H), 7.16 (1H, d, J = 2.0 Hz, Py-H), 7.03 (1H, d, J = 1.6 Hz, Py-H), 6.82 (1H, s, H-9), 4.12-4.20 (2H, m, CH2 side chain linker), 3.94 (3H, s, N-CH3), 3.88 (3H, s, 0-CH3), 3.68-3.71 (1H, m, H-11a), 3.50-3.60 (2H, m, H2-3), 2.58-2.62 (2H, m, CH2), 2.26-2.31 (4H, m, CH2), 1.50-1.54 (2H, m, CH2); 13C-NMR (125 MHz, CDCI3): δ 164.5, 162.4, 161.6, 150.5, 147.8, 140.7, 125.9, 125.5 (2C), 123.6, 123.1, 120.3, 114.6, 111.8, 111.0,94.4(2C), 68.0,63.7,56.1,53.7,51.0,46.6,36.8, 31.9, 29.6, 25.2, 24.8, 24.1, 20.2; HRMS m/z (+EI) calc, for C30H32N4O6 (M+H)+ 545.2400 found 545.2422 (M+H)+, δ 4ppm
Compound 18 was made in an analogous manner. (S)-methyl 5-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)thiophene-2-carboxylate (18). 1H-NMR (500 MHz, CDCI3): δ 7.75 (1H, s), 7.69 (2H, d, J = 8.4 Hz), 7.58 (1H, s), 7.49 (2H, d, J = 8.4), 7.23 (1H, d, J = 4.0 Hz), 7.13 (1H, d, J = 4.0 Hz), 4..24 (2H, m, CH2 side chain linker), 3.90 (3H, s, 0-CH3), 3.86 (3H, s, 0-CH3), 3.71-3.73 (1H, m, H-11 a), 3.50-3.60 (2H, m, H2-3), 2.55-2.67 (2H, m, CH2), 2.23-2.33 (4H, m, CH2), 1.56-1.74 (2H, m, CH2); 13C-NMR (125 MHz, CDCI3): δ 164.5, 162.5, 161.4, 150.6, 147.7, 140.6, 134.5, 132.9, 132.1, 131.9, 128.6, 126.8, 120.3, 114.6, 111.8, 111.0, 94.4 (2C), 68.0, 63.7, 56.1, 53.7, 52.1, 46.6, 36.8, 31.9, 29.6, 25.2, 24.8, 24.1,20.2;HRMS m/z (+EI) calc, for C29H29N3O6S (M+H)+ 547.6200 found 547.6422 (M+H)+
Example 2 (Compounds of the present invention)
General Experimental procedure for coupling different (Q1) building blocks to Alloc-THP protected PBD acid 13 to different building blocks. Example given here is for 14a. Similar procedures were followed to obtain other C8-linked Alloc-THP protected PBD-hybrid intermediates. (4-(((11S, 11aS)-10-((allyloxy)carbonyl)-7-methoxy-5-oxo-11-((tetrahydro-2H-pyran-2-yl)oxy)-2,3,5,10,11,11 a-hexahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1 -methyl-1 H-pyrrole-2-carboxylic acid (14a). A solution of Alloc-THP protected PBD acid 13 (1.85 g, 3.57 mmol, 1.2 equivalent) was dissolved in DMF. EDCI (1.24 g, 6.48 mmol, 2.0 eq) and DMAP (0.99 g, 8.1 mmol, 2.5 eq) were added to the stirred solution of 13 at room temperature and the mixture was allowed to stir for 30 minutes after which commercially available methyl 4-amino-1-methyl-1H-pyrrole-2-carboxylate (0.5 g, 3.243 mmol, 1.0 eq) was added. The reaction mixture was allowed to stir for a further 6 hour at which point TLC showed completion of reaction. The reaction was quenched by pouring it onto a mixture of ice/water mixture and the resulting mixture was extracted with ethyl acetate (3 x 150 mL). The combined extracts were sequentially washed with citric acid (200 mL), saturated aqueous NaHC03 (250 mL), water (250 mL), brine (250 mL) and finally dried over MgS04. Excess ethyl acetate was evaporated by rotary evaporator under reduced pressure and the crude product (1.88 gm) was used for hydrolysis reaction to afford 14a. For hydrolysis, Lithium hydroxide (0.24 g, 5.71 mmol, 3 eq) was added to the crude product (1.88 g, 2.87 mmol) in aqueous dioxane (75 ml dioxane, 11.5 ml water) at room temperature. The reaction mixture was stirred for 3 hours at which point TLC showed completion of reaction. Dioxane was evaporated under high vacuum and the residue was diluted with water. The resulting solution was acidified with 1 M citric acid followed by extraction with ethyl acetate (2 x 100 mL). The organic layer combined and washed with brine (100 mL), dried over MgS04 and finally concentrated using a rotary evaporator under reduced pressure to obtain 14a as a whte solid (yield, 1.68 gm, 74.0% over 2 steps) . 1H-NMR δ 9.09 (1H, s, NH), 7.39 (1H, d, J=2.0 Hz), 7.14 (1H, s, H-6), 7.12 (1H, s, H-6), 6.96 (1H, s, H-9), 6.76 (1H, d, J= 2.0 Hz, Py-H), 5.86-5.75 (2H, m, H-11), 5.13-4.84 (3H, m), 4.61-4.21 (2H,m), 4.06-3.88 (3H, m, side-chain H-1, pyran H-6), 3.87 (3H, s, 0/NCH3), 3.87 (3H, s, 0/NCH3), 3.86 (3H, s), 3.53-3.44 (3H, m), 2.55-2.45 (2H, m), 2.13-1.88 (6H, m), 1.70-1.39 (6H). m/z (+EI) calc, for C32H4oN4Oio (M)+ 640.68 found 641.57 ([M+H]+
General experimental procedure to obtain 26a from 14a bv coupling Q2 building blocks to Q1 building blocks containing intermediates (e.g., 14a). (11S, 11 aS)-allyl 7-methoxy-8-(4-((5-((4-(5-(methoxycarbonyl)-1-methyl-1H-pyrrol-3-yl)phenyl)carbamoyl)-1-methyl-1H-pyrrol-3-yl)amino)-4-oxobutoxy)-5-oxo-11-((tetrahydro-2H-pyran-2-yl)oxy)-2,3,11,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (26a). A solution of Alloc-THP protected PBD-Py acid 14a (150 mg, 0.23 mmol, 1.0 equivalent) was dissolved in DMF. EDCI (0.46 mmol, 2.0 eq) and DMAP (0.58 mmol, 2.5 eq) were added to the stirred solution of 14a at room temperature and the mixture was allowed to stir for 30 minutes after which the benzofuran-5-amine (38.3 mg, 0.29 mmol, 1.25 eq) was added. The reaction mixture was allowed to stir for a further 3 hour at which point TLC showed completion of reaction. The reaction was quenched by pouring it onto a mixture of ice/water mixture and the resulting mixture was extracted with ethyl acetate (3 x 150 ml_). The combined extracts were sequentially washed with citric acid (50 mL), saturated aqueous NaHC03 (50 mL), water (50 mL), brine (50 mL) and finally dried over MgS04. Excess ethyl acetate was evaporated by rotary evaporator under reduced pressure and the crude product was directly used in the next step without further purification. m/z (+EI) calc, for C40H45N5O10 (M)+ 755.91 found 756.76 ([M+H]+
Compounds 26a, 27a, 28a, 30a, 31a, 35a, 37a, 39a, 40a, 49a, 51a, 52a, 53a, 54a and 56a were obtained in an analogous manner.
General experimental procedure to obtain 26 from 26a bv simultaneous Alloc-THP deprotection. (i) (S)-N-(benzofuran-5-yl)-4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1 -methyl-1 H-pyrrole-2-carboxamide (26)
Palladium tetrakis[triphenylphosphine] (12.17 mg, 10.5 μΜ, 0.05 equiv) was added to a solution of of Alloc-THP-PBD conjugate 26a (154 mg, 0.21 mmol), pyrrolidine (17.91 mg, 0.25 mmol, 1.2 eq) and triphenylphosphine (13.81 mg, 0.25 equiv) in DCM (5 ml_). The reaction mixture was stirred at room temperature for 2 h at which point TLC showed completion of reaction. Excess DCM was removed by rotary evaporation under reduced pressure and the resulting residue dried in vacuo to remove pyrrolidone . The product was purified by high performance liquid chromatography (eluted with acetone:water gradient with 1% TFA) to give the product as a light yellowish solid, 26 (38 mg, 36% after HPLC purification). 1H-NMR (500 MHz, CDCI3): 7.98 (1H, s), 7.94 (1H, d, J=2.0 Hz), 7.83 (1H, s), 7.65 (1H, d, J=4.4 Hz), 7.59 (1H, d, J= 2.0 Hz), 7.52 (1H, s), 7.42 (1H, d, J= 9.0 Hz), 7.33 (1H, dd, J=8.0 Hz, 2.0 Hz), 7.10 (1H, s), 6.82 (1H, s, H-9), 6.71 (1H,s) 4.06-4.14 (2H, m, CH2), 3.90 (3H, s, A/-CH3), 3.88 (3H, s, 0-CH3), 3.66-3.72 (1H, m, H-11a) , 3.50-3.59 (2H, m, Η2-3), 2.47-2.54 (2H, m, CH2), 2.18-2.31 (4H, m, CH2), 1.95-2.08 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 169.9, 164.6, 162.7,159.9, 151.9, 150.6, 147.7, 145.7, 140.6, 133.2, 132.1, 128.5, 127.8, 123.4(2C), 121.5, 120.5, 119.6, 118.0, 113.2, 111.8, 111.3, 111.0, 106.8 (2C), 103.9, 68.1, 56.2 (2C), 53.7, 46.7(20), 36.7 (2C), 33.1, 29.8, 24.5, 24.1; HRMS m/z (+EI) calc, for C31H31N5O6 (M)+ 569.6900 found 569.6981 ([M+H]+)
Compounds 27, 28, 30, 31, 35, 37, 39, 40, 49, 51, 52, 53, 54 and 56 were obtained in an analogous manner. ii) (S)-N-(benzo[b]thiophen-5-yl)-4-(4-((7-methoxy-5-oxo-2,3,5,11a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-1 H-pyrrole-2-carboxamide (27) 1H-NMR (500 MHz, CDCI3): δ 8.23 (1H, d, J=4.5 Hz), 8.21 (1H, d, J=2.0 Hz), 7.74 (1H, d, J=8.4 Hz), 7.66 (1H, d, J= 4.0 Hz), 7.61 (1H, d, J=1.6 Hz), 7.53 (1H, d, J= 3.6 Hz), 7.48 (1H, s), 7.45 (2H, dd, J=8.0 Hz, 2.0 Hz), 7.39 (1H, s), 7.09 (1H, d, J=1.6 Hz), 6.80 (1H, s, /-/-9), 4.03-4.08 (2H, m, CH2), 3.85 (3H, s, A/-CH3), 3.82 (3H, s, O-CH3), 3.63-3.69 (1H, m, H-11a), 3.49-3.57 (2H, m, H2-3), 2.44-2.49 (2H, m, CH2), 2.15-2.29 (4H, m, CH2), 1.95-2.04 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 169.9, 164.6, 162.7,160.0, 150.7, 147.7, 140.7, 140.1, 135.1, 132.8, 131.9, 128.6, 127.3, 123.9, 123.2, 122.5, 121.6, 120.4, 119.8, 118.1, 114.8, 111.8, 110.9, 104.2, 68.1, 56.1 (2C), 53.7, 46.7(2C), 36.7 (20), 32.9, 29.5, 24.9, 24.2; iii) (S)-methyl 5-(4-(4-((7-methoxy-5-oxo-2,3,5,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-1 H-pyrrole-2-carboxamido)benzo[b]thiophene-2-carboxylate (28) 1H-NMR (500 MHz, CDCI3): δ 8.29 (1H, d, J=2.0 Hz), 8.10 (1H, s), 7.97 (1H, s), 7.76 (1H, d, J= 8.0 Hz), 7.73 (1H, s), 7.66 (1H, dd, J= 8.0 Hz, 2.0 Hz), 7.64 (1H, d, J=1.6 Hz), 7.55 (2H, dd, J=8.0 Hz, 2.0 Hz), 7.47 (2H, d, J= 3.0 Hz), 7.11 (1H, s), 6.83 (1H, s, H-9), 4.09-4.15 (2H, m, CH2), 3.93 (3H, s, A/-CH3), 3.90 (3H, s, 0-CH3), 3.87 (3H, s, O-CH3), 3.68-3.73 (1H, m, H-11a) , 3.51-3.60 (2H, m, H2-3), 2.49-2.57 (2H, m, CH2), 2.19-2.34 (4H, m, CH2), 1.94-2.09 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 169.9, 164.6, 163.2.150.6, 147.7, 139.3, 137.66, 135.7, 134.3, 132.8, 132.1, 132.0, 131.9, 130.5, 128.6, 123.1, 122.5, 121.5, 120.9, 120.0, 116.1, 111.8, 104.2, 101.1, 68.1, 56.2 (2C), 53.7, 52.5, 46.7(2C), 36.7 (2C), 33.1, 29.6, 25.0, 24.2; iv) (S)-4-(4-((7-methoxy-5-oxo-2,3,5,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-N-(4-morpholinophenyl)-1H-pyrrole-2-carboxamide (30) 1H-NMR (500 MHz, CDCI3): δ 7.71 (1H, s), 7.65 (1H, d, J=4.6 Hz), 7.52 (1H, s), 7.46 (2H, d, J= 8.4 Hz), 7.06 (1H, d, J=2.0 Hz), 6.87 (2H, d, J= 8.4 Hz), 6.82 (1H, s), 6.55 (1H, d, J=1.6 Hz), 4.05-4.19 (2H, m, CH2), 3.88 (3H, s, A/-CH3), 3.85 (3H, s, 0-CH3), 3.73-3.82 (4H, m), 3.66-3.72 (1H, m, H-11a), 3.52-3.60 (2H, m, H2-3), 3.03-3.13 (4H, m), 2.50-2.56 (2H, m, CH2), 2.19-2.33 (4H, m, CH2), 1.97-2.07 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 169.9, 164.6, 162.7, 159.6, 150.6, 148.1, 147.7, 130.8, 123.5, 121.5, 120.6, 119.5, 116.4, 111.8, 111.1, 103.8, 68.1,66.9 (2C), 56.2 (2C), 53.7, 49.8, 46.7(2C), 36.7 (2C), 33.1, 29.6, 25.0, 24.2; v) (S)-4-(4-((7-methoxy-5-oxo-2,3,5,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-N-(6-morpholinopyridin-3-yl)-1H-pyrrole-2-carboxamide (31) 1H-NMR (400 MHz, CDCI3): δ 8.23 (1H, s), 7.92 (1H, s), 7.89 (1H, d, J=8.4 Hz), 7.83 (1H, s), 7.65 (1H, d, J= 3.6 Hz), 7.51 (1H, s), 7.08 (1H, s), 6.82 (1H, s), 6.63 (1H, d, J=8.4 Hz), 6.56 (1H, s), 4.05-4.16 (2H, m, CH2), 3.91 (3H, s, A/-CH3), 3.85 (3H, s, O-CH3), 3.74-3.82 (4H, m), 3.67-3.71 (1H, m, H-11a), 3.51-3.61 (2H, m, H2-3), 3.41-3.48 (4H, m), 2.49-2.57 (2H, m, CH2), 2.17-2.26 (2H, m, CH2), 1.99-2.09 (2H, m); 13C-NMR (100 MHz, CDCI3): δ 169.9, 164.5, 162.7, 159.9, 156.8, 150.6, 147.7, 140.6, 131.5, 126.3, 123.0, 121.5, 120.6, 119.7, 111.9, 111.1, 106.8, 104.2, 68.1, 66.7 (2C), 56.2 (2C), 53.7, 46.7, 46.1(2C), 41.0, 36.7 (2C), 33.1, 29.6, 25.0, 24.2; vi) (S)-methyl 5-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-1 H-imidazole-2-carboxamido)benzo[b]thiophene-2-carboxylate (35) 1H-NMR (500 MHz, CDCI3): δ 9.04 (1H, s), 8.35 (1H, s), 8.04 (1H, s), 7.74 (1H, d, J=8.4 Hz), 7.66 (1H, d, J= 4.0 Hz), 7.61 (1H, d, J=1.6 Hz), 7.53 (1H, d,J= 3.6 Hz), 7.48 (1H, s), 7.45 (2H, dd, J=8.0 Hz, 2.0 Hz), 7.39 (1H, s), 7.09 (1H, d, J=1.6 Hz), 4.10-4.22 (2H, m, CH2), 3.96 (3H, s, A/-CH3), 3.94 (3H, s, O-CH3), 3.80 (3H, s, 0-CH3), 3.66-3.71 (1H, m, H- 11a), 3.50-3.57 (2H, m, H2-3), 2.49-2.57 (2H, m, CH2), 2.21-2.34 (4H, m, CH2), 1.95-2.08 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 169.9, 164.6, 163.2,150.6, 147.7, 139.3, 137.66, 135.7, 134.3, 132.9, 132.1, 132.0, 131.9, 130.5, 128.5, 123.2, 122.5, 121.5, 120.1, 120.0, 115.4, 114.8, 11.6, 111.0, 100.0, 67.7, 56.1 (2C), 53.7, 52.5, 46.7(2C), 35.8 (2C), 33.0, 29.6, 24.7, 24.1; vii) (S)-4-(4-((7-methoxy-5-oxo-2,3,5,11a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-N-(4-morpholinophenyl)-1H-imidazole-2-carboxamide (37) 1H-NMR (400 MHz, CDCI3): δ 8.80 (1H, s), 7.98 (1H, s), 7.83 (1H, s), 7.64 (1H, d, J = 4.0 Hz), 7.54 (1H, s), 7.51 (2H, d, J=8.4 Hz), 7.40 (1H, s), 6.90 (2H, d, J= 8.4 Hz), 6.82 (1H, s), 4.05-4.18 (2H, m, CH2), 4.01 (3H, s, A/-CH3), 3.95 (3H, s, 0-CH3), 3.74-3.82 (4H, m), 3.67-3.71 (1H, m, H-11a) , 3.52-3.57 (2H, m, H2-3), 3.11-3.14 (4H, m), 2.61-2.63 (2H, m, CH2), 2.17-2.23 (2H, m, CH2), 2.02-2.09 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 169.9, 164.5, 162.5, 156.4, 150.5, 148.3, 147.8, 140.7, 135.7, 133.9, 130.2, 120.9, 120.6, 116.3, 114.5 (2C), 111.6, 110.9, 67.8, 66.9 (2C), 56.1 (2C), 53.7, 49.7 (2C), 46.7, 35.8 (2C), 29.6, 24.8, 24.1; viii) (S)-4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-N-(4-(2-morpholinoethoxy)phenyl)-1H-imidazole-2-carboxamide (39) 1H-NMR (400 MHz, CDCI3): δ 8.80 (1H, s), 7.96 (1H, s), 7.83 (1H, s), 7.65 (1H, d, J = 4.0 Hz), 7.54 (1H, s), 7.51 (2H, d, J=8.8 Hz), 7.41 (1H, s), 6.90 (2H, d, J= 8.8 Hz), 6.83 (1H, s), 4.17-4.22 (2H, m, CH2), 4.06-4.12 (4H, m), 4.06 (3H, s, A/-CH3), 3.95 (3H, s, O-CH3), 3.72-3.79 (4H, m), 3.67-3.71 (1H, m, H-11a) , 3.52-3.57 (2H, m, H2-3), 2.79-2.81 (4H, m), 2.53-2.66 (6H, m, CH2), 2.31-2.29 (2H, m, CH2), 2.01-2.07 (2H, m); 13C-NMR (100 MHz, CDCI3): δ 174.7, 169.5, 167.0, 162.5, 155.6, 149.4, 147.8, 147.8, 140.7, 135.7, 131.8, 130.9, 121.3, 120.6, 115.1, 111.6, 110.9, 67.8,66.9(2C),66.0, 57.6, 56.1 (2C), 54.0, 53.7, 48.2 (2C), 46.7, 35.8 (2C), 33.0, 29.6, 24.8, 24.2, 24.1; ix) (S)-4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)-1-methyl-N-(6-morpholinopyridin-3-yl)-1H-imidazole-2-carboxamide (40) 1H-NMR (400 MHz, CDCI3): δ 8.73 (1H, s), 8.27 (1H, s), 7.98 (2H, m), 7.64 (1H, d, J = 3.6 Hz), 7.53 (1H, s), 7.42 (1H, s), 6.82 (1H, s), 6.64 (1H, d, J=7.2 Hz), 4.07-4.22 (2H, m, CH2), 4.04 (3H, s, A/-CH3), 3.93 (3H, s, 0-CH3), 3.78-3.85 (4H, m), 3.67-3.72 (1H, m, H-11a) , 3.52-3.59 (2H, m, H2-3), 3.44-3.5 (4H, m), 2.58-2.65 (2H, m, CH2), 2.23-2.34 (2H, m, CH2), 2.00-2.08 (2H, m); 13C-NMR (100 MHz, CDCI3): δ 169.6, 164.5, 162.7, 159.9, 150.4, 147.8, 140.6, 139.9, 135.8, 133.6, 130.4, 125.7, 120.7, 114.6, 111.7, 110.9, 106.8, 67.8, 66.7 (3C), 56.1 (2C), 53.7, 46.7, 46.1(20), 35.7 (2C), 32.9, 29.6, 24.7, 24.2; x) (S)-4-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)-1-methyl-N-(4-morpholinophenyl)-1H-pyrrole-2-carboxamide (49) 1H-NMR (400 MHz, CDCI3): δ 7.75 (1H, s), 7.9 (1H, s), 7.64 (1H, d, J=4.0 Hz), 7.52 (1H, s), 7.46 (2H, d, J= 8.0 Hz), 7.39 (2H, d, J=8.0 Hz), 7.00 (1H, s), 6.91 (1H, s), 6.90 (2H, d, J= 8.0 Hz), 6.82 (1H, s), 4.08-4.14 (2H, m, CH2), 3.98 (3H, s, A/-CH3), 3.87 (3H, s, O-CH3), 3.74-3.82 (4H, m), 3.69-3.74 (1H, m, H-11a), 3.55-3.4 (2H, m, H2-3), 3.11-3.13 (4H, m), 2.58-2.60 (2H, m, CH2), 2.25-2.31 (4H, m, CH2), 2.03-2.06 (2H, m); 13C-NMR (100 MHz, CDCI3): δ 175.4, 169.9, 162.5, 162.7, 150.4, 148.1, 147.7, 140.7, 130.8, 125.4, 124.9,123.2, 121.6, 120.4, 116.4, 111.7, 110.9, 109.3, 67.9, 66.9 (2C), 56.1 (2C), 53.7, 49.8, 46.7(2C), 36.9 (2C), 34.1, 29.6, 24.8, 24.2; xi) (S)-4-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)-1-methyl-N-(4-(2-morpholinoethoxy)phenyl)-1 H-pyrrole-2-carboxamide (51) 1H-NMR (400 MHz, CDCI3): δ 7.77 (1H, s), 7.73 (1H, s), 7.65 (1H, d, J= 4.4 Hz), 7.52 (1H, s), 7.8 (2H, d, J=8.0 Hz), 7.45 (1H, s), 7.38 (2H, d, J=8.0 Hz), 7.00 (1H, s), 6.91 (2H, d, J=8.8 Hz), 6.83 (1H, s), 4.14-4.18 (2H, m, CH2), 4.09-4.14 (4H, m), 3.98 (3H, s, A/-CH3), 3.87 (3H, s, 0-CH3), 3.77-3.83 (1H, m, H-11a), 3.71-3.75 (4H, m) , 3.52-3.57 (2H, m, Ha-3), 2.79-2.82 (2H, m), 2.53-2.64 (6H, m, CH2), 2.25-2.31 (4H, m, CH2), 2.01-2.07 (2H, m); 13C-NMR (100 MHz, CDCI3): δ 174.7, 164.6, 162.2, 159.8, 155.4, 150.5, 147.7, 140.7, 135.7, 131.8, 126.6, 125.4, 125.0, 122.0, 120.4, 115.0, 111.7, 110.9, 67.9, 66.8 (2C), 66.0, 57.6, 56.1 (2C), 54.1 (2C), 53.7, 46.7, 35.9, 33.0, 29.6, 24.8, 24.2, xii) (S)-4-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)-1-methyl-N-(4-(2-morpholinoethoxy)benzyl)-1 H-pyrrole-2-carboxamide (52) 1H-NMR (500 MHz, CDCI3): δ 7.77 (1H, s), 7.68 (1H, s), 7.65 (1H, d, J= 4.0 Hz), 7.51 (1H, s), 7.44 (2H, d, J=8.0 Hz), 7.35 (2H, d, J=8.0 Hz), 7.27 (1H, s), 6.96 (1H, d, J=2.0 Hz), 6.88 (2H, dd, J=7.0, 2.0 Hz), 6.82 (1H, s), 6.73 (1H, s), 4.50 (2H, d, J=5.5 Hz), 4.10-4.20 (4H, m, CH2), 3.98 (3H, s, A/-CH3), 3.87 (3H, s, O-CH3), 3.78-3.83 (1H, m, H-11a), 3.68-3.71 (4H, m), 3.52-3.59 (2H, m, H2-3), 2.80-2.82 (2H, m), 2.53-2.64 (6H, m, CH2), 2.24-2.33 (4H, m, CH2), 2.01-2.07 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 174.7, 164.6, 161.6, 158.1, 150.4, 147.7, 140.7, 137.5, 135.7, 131.8, 129.2, 126.6, 125.4, 124.5, 120.4, 114.9 (2C), 111.7, 110.9, 108.7, 67.9, 66.8 (2C), 66.0, 57.6, 56.1 (2C), 54.1 (2C), 53.7, 46.7, 35.9, 33.0, 29.6, 24.8, 24.2, xiii) (S)-4-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)-1-methyl-N-(6-morpholinopyridin-3-yl)-1H-pyrrole-2-carboxamide (53). 1H-NMR (500 MHz, CDCI3): δ 8.25 (1H, s), 7.96 (1H, d, J=6.5 Hz), 7.90 (1H, s), 7.85 (1H, s), 7.65 (1H, d, J= 4.0 Hz), 7.51 (1H, s), 7.44 (2H, d, J= 8.0 Hz), 7.35 (2H, d, J = 8.0 Hz), 7.00 (1H, s), 6.95 (1H, s), 6.82 (1H, s), 6.65 (1H, d, J=9.0 Hz), 4.08-4.15 (2H, m, CH2), 3.97 (3H, s, A/-CH3), 3.84 (3H, s, 0-CH3), 3.78-3.82 (4H, m), 3.67-3.70(2H, m, H2-3), 3.53-3.59 (1H, m, H-11a), 3.42-3.46 (4H, m), 2.55-2.58 (2H, m, CH2), 2.23-2.30 (4H, m, CH2), 1.99-2.05 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 174.7, 164.6, 162.7, 159.9, 156.8, 150.4, 147.7, 140.7, 131.6, 126.2, 125.4, 125.1, 120.4, 111.7, 110.8, 109.6, 106.9, 67.9, 66.7 (3C), 56.1, 53.7, 46.7, 46.1(2C), 41.0, 36.9 (2C), 34.0, 29.6, 24.8, 24.2; xiv) (S)-methyl 4-(4-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)furan-2-carboxamido)-1-methyl-1 H-pyrrole-2-carboxylate (54) 1H-NMR (500 MHz, CDCI3): δ 7.98 (1H, s, NH), 7.88 (1H, s, NH), 7.68 (1H, s, H-6), 7.65 (1H, d, J= 4.0 Hz, H-11), 7.64 (2H, d, J= 8.0 Hz, 2Ar-H), 7.54 (1H, d, J= 1.6 Hz, Py-H), 7.52 (1H, d, J= 1.6 Hz, Py-H), 7.45 (1H, d, J= 2.0 Hz, Py-H), 7.33 (2H, d, J= 8.0 Hz, 2Ar-H), 6.97 (1H, s, Py-H), 6.89 (1H, s, H-9), 4.08-4.18 (2H, m, CH2), 3.97 (3H, s, N-CH3), 3.89 (3H, s, N-CH3), 3.84 (3H, s, 0-CH3), 3.79 (3H, s, 0-CH3), 3.66-3.70 (1H, m, H-11a), 3.55-3.60 (2H, m, H2-3), 2.56-2.61 (2H, m, CH2), 2.23-2.32 (4H, m, CH2), 2.00-2.05 (2H, m); 13C-NMR (125 MHz, CDCI3): δ 162.5, 161.6, 159.1, 150.4, 147.7, 138.4, 132.8, 132.1 131.9 (2C), 128.6, 128.4 (2C), 125.4(2C), 124.8, 123.0, 121.0, 120.4 (2C), 116.2,114.6(20),109.9, 94.2, 67.4,63.6,57.1, 53.7,51.1,46.7,36.9, 36.7,34.0, 29.6, 24.2; xv) (S)-methyl 4-(4-(4-(4-(4-((7-methoxy-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)butanamido)phenyl)furan-2-carboxamido)phenyl)-1-methyl-1 H-pyrrole-2-carboxylate (56). 1H-NMR (500 MHz, CDCI3): δ 7.72 (1H, s, NH), 7.69 (1H, s, NH), 7.66 (1H, d, J= 4.0 Hz, H-11), 7.57 (2H, d, J= 8.0 Hz, 2Ar-H), 7.53 (1H, s, H-6), 7.46 (4H, d, J= 8.0 Hz, 4Ar-H), 7.41 (2H, d, J= 8.0 Hz, 2Ar-H), 7.20 (1H, d, J= 2.0 Hz, Py-H), 7.06 (1H, d, J= 2.0 Hz, Py-H), 7.02 (1H, d, J= 1.6 Hz, Py-H), 6.92 (1H, s, Py-H), 6.84 (1H, s, H-9), 4.12-4.20 (2H, m, CH2 side chain linker), 4.00 (3H, s, A/-CH3), 3.96(31-1, s, A/-CH3), 3.88 (3H, s, O-CH3), 3.84 (3H, s, 0-CH3), 3.70-3.73 (1H, m, H- 11a) , 3.55-3.61 (2H, m, H2-3), 2.58-2.62 (2H, m, CH2), 2.29-2.31 (2H, m, CH2), 1.93-2.06 (4H, m, CH2); 13C-NMR (125 MHz, CDCIs): δ 164.5, 162.4, 161.7, 150.7, 147.3, 139.2, 126.0, 125.6, 125.4 (2C), 125.2 (2C), 123.0, 120.4 (2C), 114.6 (2C), 111.4, 94.6 (2C), 68.3, 63.7, 56.1, 51.6 (2C), 41.0, 36.9, 31.9, 29.6, 25.2, 24.2, 24.1,20.2.
Example 3 - Antibacterial Evaluation
The antibacterial properties of the compounds of the invention 26, 27, 28, 30, 31, 33, 35, 37, 39, 40, 49, 51, 52, 53, 54, 56 and 59 were compared with the comparative compounds 16 and 18 against a number of bacterial strains using the CLSI broth microplate assay, as described below.
Methicillin resistant Staphylococcus aureus (MRSA) strains EMRSA-15 and EMRSA-16 were isolated from clinical material at the Royal Free Hospital, London; MRSA strain BB568 was a gift from Brigitte Berger-Bachi, Institute of Medical Microbiology, University of Zurich, Switzerland. The community-associated MRSA isolate USA300 was purchased from the American Type Culture Collection as BAA-1556. S. aureus VISA strain Mu50 is a MRSA clinical isolate with vancomycin-intermediate resistance and was isolated and provided by Keiichi Hiramatsu, Juntendo University, Tokyo. ATCC 29213 is an antibiotic susceptible S. aureus reference strain. Vancomycin resistant enterococcal isolates E. faecalisVRE1 and E. faecium VRE10 were isolated at the Royal Free Hospital. Bacteria were grown in Mueller-Hinton (MH) broth (Oxoid) or on MH agar plates at 37°C. MICs of antibacterial agents were determined by the CLSI broth microplate assay as previously described (Hadjivassileva T, Stapleton PD, Thurston DE etal. Interactions of pyrrolobenzodiazepine dimers and duplex DNA from methicillin-resistant Staphylococcus aureus. Int J Antimicrob Agents 2007; 290: 672-678); agents were dissolved in DMSO prior to dilution in broth. At the concentrations used, the solvent had no effect on bacterial growth. Three MIC determinations per strain were performed in duplicate for each compound tested.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word “comprise”, and variations such as “comprises” and “comprising”, will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

Claims (29)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
    1. A compound of formula I:
    or a salt or solvate thereof, wherein: the dotted double bond indicates the presence of a single or double bond between C2 and C3; R2 is selected from -H; =CH2; =CH(Me); C52Q aryl optionally substituted by 1 to 3 groups selected from methoxy, ethoxy, fluoro, chloro, cyano, 5 bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thienyl; C-i_5 saturated aliphatic alkyl; C3_6 saturated cycloalkyl;
    , wherein each of R21, R22 and R23 are independently selected from Η, <3ί_3 saturated alkyl, C2_3 alkenyl, C2.3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R2 group is no more than 5;
    , wherein one of R25a and R25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl and methoxy; pyridyl; and thiophenyl; and
    where R24 is selected from: H; C-i_3 saturated alkyl; C2_3 alkenyl; C2.3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl and methoxy; pyridyl; and thiophenyl; R7 is OR7Awherein R7Ais selected from Me, CH2Ph and allyl; R10 and R11 either together form a double bond, or are selected from H and QR° respectively, where Q is selected from O, S and NH and R° is H or alkyl or H and SOxM, where x is 2 or 3, and M is a monovalent pharmaceutically acceptable cation; A is selected from A1, A2, A3, A4 or A5:
    where X1 and Y1 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and O, respectively; X2 and Y2 are selected from: CH and NH; CH and NMe; N and NMe; CH and S; N and S; N and O; and CH and 0, respectively; Z1 is selected from 0 and S; Z2 is selected from CH and N; F is selected from a single bond and -(E-F1)m-; each E is independently selected from a single bond, and -C(=0)-NH-; each F1 is independently a C3.2o heteroarylene group; m is 1,2 or 3; G is selected from hydrogen, C14alkyl, -C(=0)-0-C1_4alkyl, -(CH2)n-C3.2o heterocycloalkyl, and -O-(CH2)n-C3.20 heterocycloalkyl group; each n is 0-4; provided that A2 is not A2’:
    where X1 and Y1 of A2’ are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; and provided that A3 is not A3’:
    where X2 and Y2 of A3’ are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; B is either a single bond or:
    where X and Y of B1 are selected from: CH and NMe; COH and NMe; CH and S; N and NMe; N and S, respectively; and R1 is Ci_4 alkyl.
  2. 2. A compound according to claim 1, wherein R10 and R11 form a double bond together.
  3. 3. A compound according to claim 1, wherein R10 is H and R11 is ORQ.
  4. 4. A compound according to claim 3, wherein R° is selected from H or Me.
  5. 5. A compound according to claim 1, wherein R10 is H and R11 is S03M.
  6. 6. A compound according to claim 5, wherein M is Na+.
  7. 7. A compound according to any one of claims 1 to 6, wherein R2 is of the configuration C1:
    wherein R is H or Me.
  8. 8. A compound according to any one of claims 1 to 6, wherein R2 is selected from phenyl, naphthyl, pyridyl, quinolinyl or isoquinolinyl optionally substituted by 1 to 3 groups selected from methoxy, ethoxy, fluoro, chloro, cyano, 5 bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thienyl.
  9. 9. A compound according to claim 8, wherein R2 is selected from 4-methoxy-phenyl, 3-methoxyphenyl, 4-ethoxy-phenyl, 3-ethoxy-phenyl, 4-fluoro-phenyl, 4-chloro-phenyl, 3,4-bisoxymethylene-phenyl, 4-methylthienyl, 4-cyanophenyl, 4-phenoxyphenyl, quinolin-3-yl and quinolin-6-yl, isoquinolin-3-yl and isoquinolin-6-yl, 2-thienyl, 2-furanyl, methoxynaphthyl, and naphthyl.
  10. 10. A compound according to any one of claims 1 to 6, wherein R2 is selected from methyl, ethyl, propyl, butyl or pentyl.
  11. 11. A compound according to any one of claims 1 to 6, wherein R2 is selected from cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  12. 12. A compound according to any one of claims 1 to 6, wherein R2 is
    and the total number of carbon atoms in the R2 group is no more than 4.
  13. 13. A compound according to either any one of claims 1 to 6, or claim 12, wherein one of R21, R22 and R23 is H, with the other two groups being selected from H, Ci_3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl.
  14. 14. A compound according to either any one of claims 1 to 6, or claim 12, wherein two of R21, R22 and R23 are H, with the other group being selected from H, saturated alkyl, C2-3 alkenyl, C2_3 alkynyl and cyclopropyl.
  15. 15. A compound according to either claim 13 or claim 14, wherein the groups that are not H are selected from methyl and ethyl.
  16. 16. A compound according to any one of claims 1 to 6, wherein R2 is
    and the group which is not H is phenyl optionally substituted by 1 to 3 groups selected from methoxy, ethoxy, fluoro, chloro, cyano, 5 bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thienyl.
  17. 17. A compound according to any one of claims 1 to 6, wherein R2 is and R24 is selected from H and methyl.
  18. 18. A compound according to any one of claims 1 to 6, wherein there is no double bond between C2 and C3 and R2 is H.
  19. 19. A compound according to any one of claims 1 to 18, wherein X1 and Y1 are selected from: CH and NMe; N and NMe; CH and S; and CH and O, respectively.
  20. 20. A compound according to claim 19, wherein X1 and Y1 are selected from N and NMe and CH and NMe, respectively.
  21. 21. A compound according to any one of claims 1 to 20, wherein F is -(E-F1)m-, and F1 is a C5 heteroarylene group.
  22. 22. A compound according to claim 21, wherein F1 is represented by structure F11:
    (F11) wherein X3 and Y3 are selected from: CFI and O; and CH and NMe, respectively.
  23. 23. A compound according to either claim 21 or claim 22, wherein E is a single bond.
  24. 24. A compound according to either claim 21 or claim 22, wherein E is -C(=0)-NFI-.
  25. 25. A compound according to any one of claims 1 to 24, wherein G is selected from the group consisting of-FI, -Me, -C(=0)-0-Me, -(CFI2)n-N(CFI2CFI2OCFI2CFI2), and -O-(CH2)n-N(CH2CH2OCH2CH2).
  26. 26. A compound according to any one of claims 1 to 18, wherein A is selected from the group consisting of:
  27. 27. A pharmaceutical composition comprising a compound according to any one of claims 1 to 26, and a pharmaceutically acceptable carrier or diluent.
  28. 28. Use of a compound according to any one of claims 1 to 26, in the manufacture of a medicament for the treatment of a bacterial infection.
  29. 29. A method of treatment of a patient suffering from a bacterial infection, comprising administering to said patient a therapeutically acceptable amount of a compound according to any one of claims 1 to 26, or a composition according to claim 27.
AU2013255612A 2012-04-30 2013-04-30 Pyrrolobenzodiazepines Ceased AU2013255612B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261640316P 2012-04-30 2012-04-30
US61/640,316 2012-04-30
PCT/GB2013/051097 WO2013164592A1 (en) 2012-04-30 2013-04-30 Pyrrolobenzodiazepines

Publications (2)

Publication Number Publication Date
AU2013255612A1 AU2013255612A1 (en) 2014-11-20
AU2013255612B2 true AU2013255612B2 (en) 2017-06-22

Family

ID=48289484

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013255612A Ceased AU2013255612B2 (en) 2012-04-30 2013-04-30 Pyrrolobenzodiazepines

Country Status (14)

Country Link
US (1) US9321774B2 (en)
EP (1) EP2855481A1 (en)
JP (1) JP6125614B2 (en)
KR (1) KR101960130B1 (en)
CN (1) CN104540827B (en)
AU (1) AU2013255612B2 (en)
BR (1) BR112014027190B1 (en)
CA (1) CA2872205C (en)
HK (1) HK1208217A1 (en)
IN (1) IN2014MN02175A (en)
MX (1) MX2014013144A (en)
NZ (1) NZ701478A (en)
WO (1) WO2013164592A1 (en)
ZA (1) ZA201408043B (en)

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0819095D0 (en) 2008-10-17 2008-11-26 Spirogen Ltd Pyrrolobenzodiazepines
JP5870400B2 (en) 2010-04-15 2016-03-01 シアトル ジェネティクス,インコーポレーテッド Targeted pyrrolobenzodiazepine conjugates
EP2789622B1 (en) 2010-04-15 2017-03-01 MedImmune Limited Pyrrolobenzodiazepines used to treat proliferative diseases
CN103987718A (en) 2011-09-20 2014-08-13 斯皮罗根有限公司 Pyrrolobenzodiazepines as unsymmetrical dimeric PBD compounds for inclusion in targeted conjugates
EP2751110B1 (en) 2011-10-14 2017-04-19 MedImmune Limited Asymmetrical bis-(5H-Pyrrolo[2,1-c][1,4]benzodiazepin-5-one) derivatives for the treatment of proliferative and autoimmune diseases
ES2687246T3 (en) 2011-10-14 2018-10-24 Seattle Genetics, Inc. Pyrrolobenzodiazepines and directed conjugates
SG11201401021TA (en) 2011-10-14 2014-08-28 Spirogen Sarl Pyrrolobenzodiazepines
EP2753178B1 (en) 2011-10-14 2017-12-06 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
TR201902494T4 (en) 2012-10-12 2019-03-21 Medimmune Ltd Pyrrolobenzodiazepines and their conjugates.
CA2941485C (en) 2012-10-12 2018-06-12 Philip Wilson Howard Pyrrolobenzodiazepines and conjugates thereof
CA2885305C (en) 2012-10-12 2019-11-12 Spirogen Sarl Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
SI2906298T1 (en) 2012-10-12 2018-12-31 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
CN105189507A (en) 2012-12-21 2015-12-23 斯皮罗根有限公司 Pyrrolobenzodiazepines and conjugates thereof
JP6527466B2 (en) 2012-12-21 2019-06-05 メドイミューン・リミテッドMedImmune Limited Asymmetric pyrrolobenzodiazepine dimers for use in the treatment of proliferative and autoimmune diseases
JP6340019B2 (en) 2013-03-13 2018-06-06 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepine and its conjugates
SG11201507214SA (en) 2013-03-13 2015-10-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
GB201317981D0 (en) 2013-10-11 2013-11-27 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
GB201317982D0 (en) 2013-10-11 2013-11-27 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
GB201416112D0 (en) 2014-09-12 2014-10-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
AU2015352545B2 (en) 2014-11-25 2020-10-15 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
GB201506411D0 (en) 2015-04-15 2015-05-27 Bergenbio As Humanized anti-axl antibodies
GB201506402D0 (en) 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
GB201521709D0 (en) * 2015-12-09 2016-01-20 Kings College London And Sec Dep For Health The PBD Antibacterial agents
GB201601431D0 (en) 2016-01-26 2016-03-09 Medimmune Ltd Pyrrolobenzodiazepines
GB201602356D0 (en) 2016-02-10 2016-03-23 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
GB201602359D0 (en) 2016-02-10 2016-03-23 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
GB201607478D0 (en) 2016-04-29 2016-06-15 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
WO2017201132A2 (en) 2016-05-18 2017-11-23 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
US10526294B2 (en) 2016-06-24 2020-01-07 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
GB201617466D0 (en) 2016-10-14 2016-11-30 Medimmune Ltd Pyrrolobenzodiazepine conjugates
GB201702031D0 (en) 2017-02-08 2017-03-22 Medlmmune Ltd Pyrrolobenzodiazepine-antibody conjugates
LT3544636T (en) 2017-02-08 2021-06-25 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
HUE059828T2 (en) 2017-04-18 2023-01-28 Medimmune Ltd Pyrrolobenzodiazepine conjugates
EP3612234B1 (en) 2017-04-20 2024-03-13 ADC Therapeutics SA Combination therapy with an anti-axl antibody-drug conjugate
BR112019026564A2 (en) 2017-06-14 2020-06-30 Adc Therapeutics Sa dosing regimens for administration of an anti-cd19 adc
AU2018316532B2 (en) 2017-08-18 2022-11-24 Medimmune Limited Pyrrolobenzodiazepine conjugates
EP3690038A4 (en) 2017-09-29 2021-05-19 Daiichi Sankyo Company, Limited Antibody-pyrrolobenzodiazepine derivative conjugate
EP3717021A1 (en) 2017-11-27 2020-10-07 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
GB201803342D0 (en) 2018-03-01 2018-04-18 Medimmune Ltd Methods
GB201806022D0 (en) 2018-04-12 2018-05-30 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
GB201900929D0 (en) * 2019-01-23 2019-03-13 King S College London Nf-kappa b inhibitors
BR112022013008A2 (en) 2019-12-31 2022-09-06 Legochem Biosciences Inc PYRROLOBENZODIAZEPINE DERIVATIVE COMPOUND, PYRROLOBENZODIAZEPINE BINDING DERIVATIVE CONJUGATE, AND, PHARMACEUTICAL COMPOSITION AND METHOD FOR PREVENTION OR TREATMENT OF PROLIFERATIVE DISEASE
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005032594A2 (en) * 2003-10-03 2005-04-14 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Alkylators linked to polyamides as dna binding agents
WO2007039752A1 (en) * 2005-10-05 2007-04-12 Spirogen Limited Alkyl 4- [4- (5-oxo-2, 3, 5, 11a-tetrahyd0-5h-pyrr0l0 [2, 1-c] [1, 4] benzodiazepine-8-yloxy) -butyrylamino]-1h-pyrrole-2-carboxylate derivatives and related compounds for the treatment of a proliferative disease
WO2009060208A1 (en) * 2007-11-09 2009-05-14 Spirogen Limited Pyrrolobenzodiazepines

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5569587A (en) 1978-11-17 1980-05-26 Meiji Seika Kaisha Ltd Novel antibiotic neothramycin derivative and its preparation
JPS5615289A (en) 1979-07-17 1981-02-14 Green Cross Corp:The Novel benzodiazepinnbased compound 3
JPS58180487A (en) 1982-04-16 1983-10-21 Kyowa Hakko Kogyo Co Ltd Antibiotic dc-81 and its preparation
US5229490A (en) 1987-05-06 1993-07-20 The Rockefeller University Multiple antigen peptide system
ATE236890T1 (en) 1996-12-23 2003-04-15 Bristol Myers Squibb Pharma Co OXYGEN OR SULFUR CONTAINING 5-MEMBED HETEROAROMATIC DERIVATIVES AS FACTOR XA INHIBITORS
US6187797B1 (en) 1996-12-23 2001-02-13 Dupont Pharmaceuticals Company Phenyl-isoxazoles as factor XA Inhibitors
AU2713799A (en) 1998-03-12 1999-09-27 Novo Nordisk A/S Modulators of protein tyrosine phosphatases (ptpases)
GB9818732D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Collection of compounds
GB9818730D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Collections of compounds
GB9818731D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Compounds
DE69930328T2 (en) 1998-08-27 2006-12-14 Spirogen Ltd., Ryde Dimer pyrrolobenzodiazepines
US6909006B1 (en) 1999-08-27 2005-06-21 Spirogen Limited Cyclopropylindole derivatives
BR0016565A (en) 1999-12-20 2002-09-17 Univ North Carolina Diamidine compounds as small DNA groove ligands
WO2002062755A2 (en) * 2000-12-27 2002-08-15 Genelabs Technologies, Inc. Polyamide analogs as dna minor groove binders
JP2005505507A (en) * 2001-06-13 2005-02-24 ジーンソフト ファーマシューティカルズ インコーポレイテッド Isoquinoline compounds having anti-infective activity
EP1423110A4 (en) * 2001-09-07 2005-04-27 Scripps Research Inst Cbi analogues of cc-1065 and the duocarmycins
US20040138269A1 (en) 2002-10-11 2004-07-15 Sugen, Inc. Substituted pyrroles as kinase inhibitors
GB0226593D0 (en) 2002-11-14 2002-12-24 Consultants Ltd Compounds
GB0321295D0 (en) 2003-09-11 2003-10-15 Spirogen Ltd Synthesis of protected pyrrolobenzodiazepines
GB0404577D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Pyrrolobenzodiazepines
GB0404578D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Pyrrolobenzodiazepines
GB0404574D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Amino acids
US7741319B2 (en) 2004-03-01 2010-06-22 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
US7528126B2 (en) 2004-03-09 2009-05-05 Spirogen Limited Pyrrolobenzodiazepines
GB0410725D0 (en) 2004-05-13 2004-06-16 Spirogen Ltd Pyrrolobenzodiazepine therapeutic agents
PT1879901E (en) 2005-04-21 2010-03-29 Spirogen Ltd Pyrrolobenzodiazepines
WO2008060208A1 (en) 2006-11-16 2008-05-22 Telefonaktiebolaget L M Ericsson (Publ) Gateway selection mechanism
GB0813432D0 (en) 2008-07-22 2008-08-27 Spirogen Ltd Pyrrolobenzodiazepines
GB0819095D0 (en) 2008-10-17 2008-11-26 Spirogen Ltd Pyrrolobenzodiazepines

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005032594A2 (en) * 2003-10-03 2005-04-14 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Alkylators linked to polyamides as dna binding agents
WO2007039752A1 (en) * 2005-10-05 2007-04-12 Spirogen Limited Alkyl 4- [4- (5-oxo-2, 3, 5, 11a-tetrahyd0-5h-pyrr0l0 [2, 1-c] [1, 4] benzodiazepine-8-yloxy) -butyrylamino]-1h-pyrrole-2-carboxylate derivatives and related compounds for the treatment of a proliferative disease
WO2009060208A1 (en) * 2007-11-09 2009-05-14 Spirogen Limited Pyrrolobenzodiazepines

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BRUCOLI, F. et al, Bioorganic & Medicinal Chemistry Letters, 2011, vol. 21, pp. 3780-3783 *
KAMAL, A. et al, Bioorganic & Medicinal Chemistry, 2010, vol. 18, pp. 4747-4761 *
KAMAL, A. et al, Bioorganic & Medicinal Chemistry, 2010, vol. 18, pp. 6666-6677 *
RAHMAN, K.M. et al, Chemical Communications, 2009, pp. 4079-4099 *
RAHMAN, K.M. et al, Journal of Antimicrobial Chemotherapy, 2012, vol. 67, pp. 1683-1696 *

Also Published As

Publication number Publication date
KR101960130B1 (en) 2019-03-19
JP6125614B2 (en) 2017-05-10
CA2872205A1 (en) 2013-11-07
CN104540827B (en) 2018-03-23
JP2015516987A (en) 2015-06-18
US20150126495A1 (en) 2015-05-07
BR112014027190B1 (en) 2020-03-03
MX2014013144A (en) 2015-05-11
EP2855481A1 (en) 2015-04-08
CN104540827A (en) 2015-04-22
US9321774B2 (en) 2016-04-26
KR20150014935A (en) 2015-02-09
WO2013164592A1 (en) 2013-11-07
AU2013255612A1 (en) 2014-11-20
ZA201408043B (en) 2017-08-30
HK1208217A1 (en) 2016-02-26
NZ701478A (en) 2016-08-26
IN2014MN02175A (en) 2015-08-28
BR112014027190A2 (en) 2017-06-27
CA2872205C (en) 2020-07-21

Similar Documents

Publication Publication Date Title
AU2013255612B2 (en) Pyrrolobenzodiazepines
US8940733B2 (en) Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
EP1723152B1 (en) Pyrrolobenzodiazepines
AU2003282251B2 (en) Pyrrolobenzodiazepines
KR101960129B1 (en) Pyrrolobenzodiazepines
US8637664B2 (en) Alkyl 4- [4- (5-oxo-2,3,5, 11a-tetrahydo-5H-pyrrolo [2, 1-c] [1,4] benzodiazepine-8-yloxy)-butyrylamino]-1H-pyrrole-2-carboxylate derivatives and related compounds for the treatment of a proliferative disease
NZ563136A (en) Pyrrolobenzodiazepines
WO2009060208A1 (en) Pyrrolobenzodiazepines

Legal Events

Date Code Title Description
PC1 Assignment before grant (sect. 113)

Owner name: UCL BUSINESS PLC; MEDIMMUNE LIMITED

Free format text: FORMER APPLICANT(S): SPIROGEN SARL; UCL BUSINESS PLC

PC1 Assignment before grant (sect. 113)

Owner name: MEDIMMUNE LIMITED

Free format text: FORMER APPLICANT(S): MEDIMMUNE LIMITED; UCL BUSINESS PLC

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired